1
|
Xu J, Ruan X. Schwann cell autotransplantation for the treatment of peripheral nerve injury. Life Sci 2024; 358:123129. [PMID: 39393574 DOI: 10.1016/j.lfs.2024.123129] [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: 03/08/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Peripheral nerve injury occurs in a relatively large proportion of trauma patients, in whom it generally results in severe functional impairment and permanent disability. At present, however, there are no effective treatments available. Studies have shown that Schwann cells play an indispensable role in removing myelin debris and guiding axonal regeneration, and transplantation using autologous Schwann cells has shown good efficacy for patients with peripheral nerve injury. In recent years, Schwann cell autologous transplantation therapy has become an area of intensive research and is anticipated to provide a new strategy for the clinical treatment of peripheral nerve injury. In this article, we review the rationale for selecting Schwann cell autotransplantation therapy and the latest progress in key aspects of cell transplantation and clinical efficacy, and also summarize the future directions of research on this therapy. All of the above provide a strong basis for the further improvement and clinical promotion of this therapy.
Collapse
Affiliation(s)
- Jialiang Xu
- China Medical University, Shenyang, Liaoning 110122, People's Republic of China.
| | - Xuelei Ruan
- Department of Neurobiology, China Medical University, Shenyang, Liaoning 110122, People's Republic of China.
| |
Collapse
|
2
|
Tusnim J, Kutuzov P, Grasman JM. In Vitro Models for Peripheral Nerve Regeneration. Adv Healthc Mater 2024; 13:e2401605. [PMID: 39324286 DOI: 10.1002/adhm.202401605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/14/2024] [Indexed: 09/27/2024]
Abstract
Peripheral nerve injury (PNI) resulting in lesions is highly prevalent clinically, but current therapeutic approaches fail to provide satisfactory outcomes in many patients. While peripheral nerves have intrinsic regenerative capacity, the regenerative capabilities of peripheral nerves are often insufficient to restore full functionality. This highlights an unmet need for developing more effective strategies to repair damaged peripheral nerves and improve regenerative success. Consequently, researchers are actively exploring a variety of therapeutic strategies, encompassing the local delivery of trophic factors or bioactive molecules, the design of advanced biomaterials that interact with regenerating axons, and augmentation with nerve guidance conduits or complex prostheses. However, clinical translation of these technologies remains limited, emphasizing the need for continued research on peripheral nerve regeneration modalities that can enhance functional restoration. Experimental models that accurately recapitulate key aspects of peripheral nerve injury and repair biology can accelerate therapeutic development by enabling systematic testing of new techniques. Advancing regenerative therapies for PNI requires bridging the gap between basic science discoveries and clinical application. This review discusses different in vitro models of peripheral nerve injury and repair, including their advantages, limitations, and potential applications.
Collapse
Affiliation(s)
- Jarin Tusnim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Peter Kutuzov
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jonathan M Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| |
Collapse
|
3
|
Duvernay J, Gengler C, Le Van T, Daubail B, di Summa P, Zwetyenga N, Guillier D. In vivo evaluation of Adipose-Derived Stem Cells (ADSCs) using Nanofat technique and chitosan conduit for peripheral nerve defect repair. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2023; 124:101491. [PMID: 37150480 DOI: 10.1016/j.jormas.2023.101491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/29/2023] [Accepted: 05/03/2023] [Indexed: 05/09/2023]
Abstract
INTRODUCTION Nerve autograft is the gold standard for reconstruction of peripheral nerve loss. The alternative is the interposition of a synthetic regeneration conduit. The purpose of the study was to evaluate the in vivo feasibility and interest of Adipose-Derived Stem Cells (ADSCs) using the Nanofat technique and chitosan conduit for peripheral nerve defect repair. MATERIAL AND METHODS In vivo, after the creation of a 10 mm defect of the sciatic nerve, 2 groups were defined according to the nerve repair in rats: "chitosan" group (n = 10) and "chitosan and ADSCs" group (n = 10) with a clinical and paraclinical evaluation at 7 weeks. RESULTS The in vivo results seem to show that the adjunction of ADSCs was favorable clinically, histologically and functionally compare to a chitosan reconstruction alone. DISCUSSION Peripheral nerve repair with defect using a chitosan conduit associated with ADSCs would constitute a surgical alternative in a single surgical step.
Collapse
Affiliation(s)
- Justine Duvernay
- Oral and Maxillofacial department (CHU Bordeaux, France), Centre Francois Xavier Michelet Groupe Hospitalier Pellegrin Place Amélie Raba Léon 33000 Bordeaux, France.
| | - Charline Gengler
- Oral and Maxillofacial department (CHU Dijon, France), 14 Rue Paul Gaffarel, 21000 Dijon, France
| | - Tuan Le Van
- Neurosurgery department (CHU Dijon, France), 14 Rue Paul Gaffarel, 21000 Dijon, France
| | - Benoit Daubail
- Neurology department (CHU Dijon, France), 14 Rue Paul Gaffarel, 21000 Dijon, France
| | - Pietro di Summa
- Plastic and reconstructive department (CHUV Lausanne, Suisse), Rue du Bugnon 46, 1011 Lausanne, Suisse
| | - Narcisse Zwetyenga
- Oral and Maxillofacial department (CHU Dijon, France), 14 Rue Paul Gaffarel, 21000 Dijon, France
| | - David Guillier
- Plastic and reconstructive department (CHU Dijon, France), 14 Rue Paul Gaffarel, 21000 Dijon, France
| |
Collapse
|
4
|
Brambilla S, Guiotto M, Torretta E, Armenia I, Moretti M, Gelfi C, Palombella S, di Summa PG. Human platelet lysate stimulates neurotrophic properties of human adipose-derived stem cells better than Schwann cell-like cells. Stem Cell Res Ther 2023; 14:179. [PMID: 37480149 PMCID: PMC10362751 DOI: 10.1186/s13287-023-03407-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/06/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Trauma-associated peripheral nerve injury is a widespread clinical problem causing sensory and motor disabilities. Schwann cells (SCs) contribute to nerve regeneration, mainly by secreting nerve growth factor (NGF) and brain-derived neurotrophic factor. In the last years, adipose-derived stem cells (ASCs) differentiated into SCs (SC-ASCs) were considered as promising cell therapy. However, the cell trans-differentiation process has not been effectively showed and presents several drawbacks, thus an alternative approach for increasing ASCs neurotrophic properties is highly demanded. In the context of human cell-based therapies, Good Manufacturing Practice directions indicate that FBS should be substituted with a xenogeneic-free supplement, such as Human Platelet Lysate (HPL). Previously, we demonstrated that neurotrophic properties of HPL-cultured ASCs were superior compared to undifferentiated FBS-cultured ASCs. Therefore, as following step, here we compared the neurotrophic properties of differentiated SC-like ASCs and HPL-cultured ASCs. METHODS Both cell groups were investigated for gene expression level of neurotrophic factors, their receptors and neuronal markers. Moreover, the expression of nestin was quantitatively evaluated by flow cytometry. The commitment toward the SC phenotype was assessed with immunofluorescence pictures. Proteomics analysis was performed on both cells and their conditioned media to compare the differential protein profile. Finally, neurotrophic abilities of both groups were evaluated with a functional co-culture assay, assessing dorsal root ganglia survival and neurite outgrowth. RESULTS HPL-cultured ASCs demonstrated higher gene expression of NGF and lower expression of S100B. Moreover, nestin was present in almost all HPL-cultured ASCs and only in one quarter of SC-ASCs. Immunofluorescence confirmed that S100B was not present in HPL-cultured ASCs. Proteomics analysis validated the higher expression of nestin and the increase in cytoskeletal and ECM proteins involved in neural regeneration processes. The co-culture assay highlighted that neurite outgrowth was higher in the presence of HPL-ASCs or their conditioned medium compared to SC-ASCs. CONCLUSIONS All together, our results show that HPL-ASCs were more neurotrophic than SC-ASCs. We highlighted that the HPL triggers an immature neuro-induction state of ASCs, while keeping their stem properties, paving the way for innovative therapies for nerve regeneration.
Collapse
Affiliation(s)
- Stefania Brambilla
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via C. Belgioioso 173, 20157, Milan, Italy
| | - Martino Guiotto
- Department of Plastic and Hand Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Enrica Torretta
- Laboratory of Proteomics and Lipidomics, IRCCS Istituto Ortopedico Galeazzi, Via C. Belgioioso 173, 20157, Milan, Italy
| | - Ilaria Armenia
- Instituto de Nanociencia y Materiales de Aragón, CSIC-University of Zaragoza, C/ Pedro Cerbuna 12, 50009, Zaragoza, Spain
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via C. Belgioioso 173, 20157, Milan, Italy
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Via F. Chiesa 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, USI, Lugano, Switzerland
| | - Cecilia Gelfi
- Laboratory of Proteomics and Lipidomics, IRCCS Istituto Ortopedico Galeazzi, Via C. Belgioioso 173, 20157, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Silvia Palombella
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via C. Belgioioso 173, 20157, Milan, Italy.
| | - Pietro G di Summa
- Department of Plastic and Hand Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
5
|
Ferreira LVDO, Kamura BDC, de Oliveira JPM, Chimenes ND, de Carvalho M, dos Santos LA, Dias-Melicio LA, Amorim RL, Amorim RM. In Vitro Transdifferentiation Potential of Equine Mesenchymal Stem Cells into Schwann-Like Cells. Stem Cells Dev 2023; 32:422-432. [PMID: 37071193 PMCID: PMC10401561 DOI: 10.1089/scd.2022.0274] [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: 11/22/2022] [Accepted: 04/17/2023] [Indexed: 04/19/2023] Open
Abstract
Schwann cells (SCs) are essential for the regenerative processes of peripheral nerve injuries. However, their use in cell therapy is limited. In this context, several studies have demonstrated the ability of mesenchymal stem cells (MSCs) to transdifferentiate into Schwann-like cells (SLCs) using chemical protocols or co-culture with SCs. Here, we describe for the first time the in vitro transdifferentiation potential of MSCs derived from equine adipose tissue (AT) and equine bone marrow (BM) into SLCs using a practical method. In this study, the facial nerve of a horse was collected, cut into fragments, and incubated in cell culture medium for 48 h. This medium was used to transdifferentiate the MSCs into SLCs. Equine AT-MSCs and BM-MSCs were incubated with the induction medium for 5 days. After this period, the morphology, cell viability, metabolic activity, gene expression of glial markers glial fibrillary acidic protein (GFAP), myelin basic protein (MBP), p75 and S100β, nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and glial cell-derived neurotrophic factor (GDNF), and the protein expression of S100 and GFAP were evaluated in undifferentiated and differentiated cells. The MSCs from the two sources incubated with the induction medium exhibited similar morphology to the SCs and maintained cell viability and metabolic activity. There was a significant increase in the gene expression of BDNF, GDNF, GFAP, MBP, p75, and S100β in equine AT-MSCs and GDNF, GFAP, MBP, p75, and S100β in equine BM-MSCs post-differentiation. Immunofluorescence analysis revealed GFAP expression in undifferentiated and differentiated cells, with a significant increase in the integrated pixel density in differentiated cells and S100 was only expressed in differentiated cells from both sources. These findings indicate that equine AT-MSCs and BM-MSCs have great transdifferentiation potential into SLCs using this method, and they represent a promising strategy for cell-based therapy for peripheral nerve regeneration in horses.
Collapse
Affiliation(s)
- Lucas Vinícius de Oliveira Ferreira
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Translational Nucleus of Regenerative Medicine (NUTRAMERE), School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Beatriz da Costa Kamura
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Translational Nucleus of Regenerative Medicine (NUTRAMERE), School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - João Pedro Marmol de Oliveira
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Translational Nucleus of Regenerative Medicine (NUTRAMERE), School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Natielly Dias Chimenes
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Translational Nucleus of Regenerative Medicine (NUTRAMERE), School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Márcio de Carvalho
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Leandro Alves dos Santos
- Confocal Microscopy Laboratory, UNIPEX–Experimental Research Unit, Medical School of Botucatu; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Luciane Alarcão Dias-Melicio
- Confocal Microscopy Laboratory, UNIPEX–Experimental Research Unit, Medical School of Botucatu; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Department of Pathology, Medical School of Botucatu; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Renée Laufer Amorim
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Rogério Martins Amorim
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Translational Nucleus of Regenerative Medicine (NUTRAMERE), School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
6
|
Supra R, Agrawal DK. Peripheral Nerve Regeneration: Opportunities and Challenges. JOURNAL OF SPINE RESEARCH AND SURGERY 2023; 5:10-18. [PMID: 36873243 PMCID: PMC9983644 DOI: 10.26502/fjsrs0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Peripheral nerve injury has detrimental effects on the quality of life for patients and is a worldwide issue with high rates of morbidity. Research on the molecular mechanisms of nerve injury, microsurgical techniques, and advances in stem cell research have led to substantial progress in the field of translational neurophysiology. Current research on peripheral nerve regeneration aims to accelerate peripheral nerve development through pluripotent stem cells and potential use of smart exosomes, pharmacological agents, and bioengineering of nerve conduits. In this article critically reviewed and summarized various methods used for peripheral nerve regeneration and highlight the opportunities and challenges that come along with these strategies.
Collapse
Affiliation(s)
- Rajiv Supra
- College of Osteopathic Medicine, Touro University, Henderson, Nevada
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Pomona, California
| |
Collapse
|
7
|
Secretome of Mesenchymal Stromal Cells as a Possible Innovative Therapeutic Tool in Facial Nerve Injury Treatment. BIOMED RESEARCH INTERNATIONAL 2023; 2023:8427200. [PMID: 36691473 PMCID: PMC9867597 DOI: 10.1155/2023/8427200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Facial nerve palsy is a serious neurological condition that strongly affects patient everyday life. Standard treatments provide insufficient improvement and are burdened with the risk of severe complications, e.g., facial synkinesis. Mesenchymal stromal cell-based therapies are a novel and extensively developed field which offers new treatment approaches with promising results in regards to the nervous tissue regeneration. The potential of mesenchymal stromal cells (MSCs) to aid the regeneration of damaged nerves has been demonstrated in several preclinical models, as well as in several clinical trials. However, therapies based on cell transplantation are difficult to standardize in the manner similar to that of routine clinical practices. On the other hand, treatments based on mesenchymal stromal cell secretome harness the proregenerative features of mesenchymal stromal cells but relay on a product with measurable parameters that can be put through standardization procedures and deliver a fully controllable end-product. Utilization of mesenchymal stromal cell secretome allows the controlled dosage and standardization of the components to maximize the therapeutic potential and ensure safety of the end-product.
Collapse
|
8
|
Hu T, Chang S, Qi F, Zhang Z, Chen J, Jiang L, Wang D, Deng C, Nie K, Xu G, Wei Z. Neural grafts containing exosomes derived from Schwann cell-like cells promote peripheral nerve regeneration in rats. BURNS & TRAUMA 2023; 11:tkad013. [PMID: 37122841 PMCID: PMC10141455 DOI: 10.1093/burnst/tkad013] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/06/2022] [Accepted: 03/02/2023] [Indexed: 05/02/2023]
Abstract
Background Schwann cell-like cells (SCLCs), differentiated from mesenchymal stem cells, have shown promising outcomes in the treatment of peripheral nerve injuries in preclinical studies. However, certain clinical obstacles limit their application. Hence, the primary aim of this study was to investigate the role of exosomes derived from SCLCs (SCLCs-exo) in peripheral nerve regeneration. Methods SCLCs were differentiated from human amniotic mesenchymal stem cells (hAMSCs) in vitro and validated by immunofluorescence, real-time quantitative PCR and western blot analysis. Exosomes derived from hAMSCs (hAMSCs-exo) and SCLCs were isolated by ultracentrifugation and validated by nanoparticle tracking analysis, WB analysis and electron microscopy. A prefabricated nerve graft was used to deliver hAMSCs-exo or SCLCs-exo in an injured sciatic nerve rat model. The effects of hAMSCs-exo or SCLCs-exo on rat peripheral nerve injury (PNI) regeneration were determined based on the recovery of neurological function and histomorphometric variation. The effects of hAMSCs-exo or SCLCs-exo on Schwann cells were also determined via cell proliferation and migration assessment. Results SCLCs significantly expressed the Schwann cell markers glial fibrillary acidic protein and S100. Compared to hAMSCs-exo, SCLCs-exo significantly enhanced motor function recovery, attenuated gastrocnemius muscle atrophy and facilitated axonal regrowth, myelin formation and angiogenesis in the rat model. Furthermore, hAMSCs-exo and SCLCs-exo were efficiently absorbed by Schwann cells. However, compared to hAMSCs-exo, SCLCs-exo significantly promoted the proliferation and migration of Schwann cells. SCLCs-exo also significantly upregulated the expression of a glial cell-derived neurotrophic factor, myelin positive regulators (SRY-box transcription factor 10, early growth response protein 2 and organic cation/carnitine transporter 6) and myelin proteins (myelin basic protein and myelin protein zero) in Schwann cells. Conclusions These findings suggest that SCLCs-exo can more efficiently promote PNI regeneration than hAMSCs-exo and are a potentially novel therapeutic approach for treating PNI.
Collapse
Affiliation(s)
| | | | - Fang Qi
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Zhonghui Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Jiayin Chen
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Lingli Jiang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Dali Wang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Chengliang Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Kaiyu Nie
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | | | - Zairong Wei
- Correspondence. Guangchao Xu, ; Zairong Wei,
| |
Collapse
|
9
|
Yousefifard M, Sarveazad A, Janzadeh A, Behroozi Z, Nasirinezhad F. Pain Alleviating Effect of Adipose-Derived Stem Cells Transplantation on the Injured Spinal Cord: A Behavioral and Electrophysiological Evaluation. J Stem Cells Regen Med 2022; 18:53-63. [PMID: 36713791 PMCID: PMC9837693 DOI: 10.46582/jsrm.1802010] [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: 05/18/2022] [Accepted: 09/15/2022] [Indexed: 01/01/2023]
Abstract
Few studies are conducted on the efficacy of human adipose-derived stem cells (ADSCs) in spinal cord injury (SCI) management and electrophysiological changes in the spinal cord. Therefore, the present study aimed to determine the effect of ADSCs on neuropathic pain, motor function recovery, and electrophysiology assessment. For the purpose of this study, adult male Wistar rats (weight: 140-160 gr, n = 42) were randomly allocated into five groups namely intact animals, sham-operated, SCI non-treated animals, vehicle-treated (culture media), and ADSCs treated groups. One week after clips compression SCI induction, about 1×106 cells were transplanted into the spinal cord. As well, both neuropathic pain (allodynia and hyperalgesia) and motor function were measured weekly. Cavity size, ADSCs survival, and electrophysiology assessments were measured at the end of the eighth week. The transplantation of ADSCs resulted in a significant improvement in the locomotion of SCI animals (p<0.0001), mechanical allodynia (p<0.0001), cold allodynia (p<0.0001), mechanical hyperalgesia (p<0.0001), and thermal hyperalgesia (p<0.0001). The cavity size was significantly smaller among the ADSCs-treated animals (p <0.0001). The single-unit recording showed that the transplantation of ADSCs decreased wide dynamic range (WDR) in neurons and it evoked potential in response to receiving signals from Aβ (p<0.0001) and Aδ (p=0.003) C-fiber (p<0.0001) neurons. Post-discharge recorded from WDR neurons decreased after the transplantation of ADSCs (p<0.0001) and wind up in the ADSCs-treated group was lower than that of the SCI group (p=0.003). Our results showed that the transplantation of ADSCs could significantly alleviate neuropathic pain, enhance motor function recovery, and improve electrophysiology findings after SCI.
Collapse
Affiliation(s)
- Mahmoud Yousefifard
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Arash Sarveazad
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran,Nursing care Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Atousa Janzadeh
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Behroozi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Farinaz Nasirinezhad
- Cellular and molecular research center, Iran University of Medical Sciences, Tehran, Iran,Centre for Experimental and Comparative Study, Iran University of Medical Sciences, Tehran, Iran,Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran,Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran,Farinaz Nasirinezhad, Cellular, and molecular research center, Iran University of Medical Sciences, Tehran, Iran., Tel/Fax: +982188622709.
| |
Collapse
|
10
|
Hörner SJ, Couturier N, Gueiber DC, Hafner M, Rudolf R. Development and In Vitro Differentiation of Schwann Cells. Cells 2022; 11:3753. [PMID: 36497014 PMCID: PMC9739763 DOI: 10.3390/cells11233753] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Schwann cells are glial cells of the peripheral nervous system. They exist in several subtypes and perform a variety of functions in nerves. Their derivation and culture in vitro are interesting for applications ranging from disease modeling to tissue engineering. Since primary human Schwann cells are challenging to obtain in large quantities, in vitro differentiation from other cell types presents an alternative. Here, we first review the current knowledge on the developmental signaling mechanisms that determine neural crest and Schwann cell differentiation in vivo. Next, an overview of studies on the in vitro differentiation of Schwann cells from multipotent stem cell sources is provided. The molecules frequently used in those protocols and their involvement in the relevant signaling pathways are put into context and discussed. Focusing on hiPSC- and hESC-based studies, different protocols are described and compared, regarding cell sources, differentiation methods, characterization of cells, and protocol efficiency. A brief insight into developments regarding the culture and differentiation of Schwann cells in 3D is given. In summary, this contribution provides an overview of the current resources and methods for the differentiation of Schwann cells, it supports the comparison and refinement of protocols and aids the choice of suitable methods for specific applications.
Collapse
Affiliation(s)
- Sarah Janice Hörner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Nathalie Couturier
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Daniele Caroline Gueiber
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Department of Electronics Engineering, Federal University of Technology Paraná, Ponta Grossa 84017-220, Brazil
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| |
Collapse
|
11
|
Podder AK, Mohamed MA, Tseropoulos G, Nasiri B, Andreadis ST. Engineering Nanofiber Scaffolds with Biomimetic Cues for Differentiation of Skin-Derived Neural Crest-like Stem Cells to Schwann Cells. Int J Mol Sci 2022; 23:10834. [PMID: 36142746 PMCID: PMC9504850 DOI: 10.3390/ijms231810834] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/31/2022] [Accepted: 09/13/2022] [Indexed: 01/17/2023] Open
Abstract
Our laboratory reported the derivation of neural crest stem cell (NCSC)-like cells from the interfollicular epidermis of the neonatal and adult epidermis. These keratinocyte (KC)-derived Neural Crest (NC)-like cells (KC-NC) could differentiate into functional neurons, Schwann cells (SC), melanocytes, and smooth muscle cells in vitro. Most notably, KC-NC migrated along stereotypical pathways and gave rise to multiple NC derivatives upon transplantation into chicken embryos, corroborating their NC phenotype. Here, we present an innovative design concept for developing anisotropically aligned scaffolds with chemically immobilized biological cues to promote differentiation of the KC-NC towards the SC. Specifically, we designed electrospun nanofibers and examined the effect of bioactive cues in guiding KC-NC differentiation into SC. KC-NC attached to nanofibers and adopted a spindle-like morphology, similar to the native extracellular matrix (ECM) microarchitecture of the peripheral nerves. Immobilization of biological cues, especially Neuregulin1 (NRG1) promoted the differentiation of KC-NC into the SC lineage. This study suggests that poly-ε-caprolactone (PCL) nanofibers decorated with topographical and cell-instructive cues may be a potential platform for enhancing KC-NC differentiation toward SC.
Collapse
Affiliation(s)
- Ashis Kumar Podder
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
- School of Pharmacy, Brac University, Dhaka 1212, Bangladesh
| | - Mohamed Alaa Mohamed
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
- Department of Chemistry, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Georgios Tseropoulos
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
| | - Bita Nasiri
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
| | - Stelios T. Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York (SUNY); Buffalo, NY 14260, USA
- Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
- Center of Cell, Gene and Tissue Engineering (CGTE), University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
| |
Collapse
|
12
|
Liu B, Kong Y, Shi W, Kuss M, Liao K, Hu G, Xiao P, Sankarasubramanian J, Guda C, Wang X, Lei Y, Duan B. Exosomes derived from differentiated human ADMSC with the Schwann cell phenotype modulate peripheral nerve-related cellular functions. Bioact Mater 2022; 14:61-75. [PMID: 35310346 PMCID: PMC8892082 DOI: 10.1016/j.bioactmat.2021.11.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/09/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Peripheral nerve regeneration remains a significant clinical challenge due to the unsatisfactory functional recovery and public health burden. Exosomes, especially those derived from mesenchymal stem cells (MSCs), are promising as potential cell-free therapeutics and gene therapy vehicles for promoting neural regeneration. In this study, we reported the differentiation of human adipose derived MSCs (hADMSCs) towards the Schwann cell (SC) phenotype (hADMSC-SCs) and then isolated exosomes from hADMSCs with and without differentiation (i.e., dExo vs uExo). We assessed and compared the effects of uExo and dExo on antioxidative, angiogenic, anti-inflammatory, and axon growth promoting properties by using various peripheral nerve-related cells. Our results demonstrated that hADMSC-SCs secreted more neurotrophic factors and other growth factors, compared to hADMSCs without differentiation. The dExo isolated from hADMSC-SCs protected rat SCs from oxidative stress and enhanced HUVEC migration and angiogenesis. Compared to uExo, dExo also had improved performances in downregulating pro-inflammatory gene expressions and cytokine secretions and promoting axonal growth of sensory neurons differentiated from human induced pluripotent stem cells. Furthermore, microRNA (miRNA) sequencing analysis revealed that exosomes and their parent cells shared some similarities in their miRNA profiles and exosomes displayed a distinct miRNA signature. Many more miRNAs were identified in dExo than in uExo. Several upregulated miRNAs, like miRNA-132-3p and miRNA-199b-5p, were highly related to neuroprotection, anti-inflammation, and angiogenesis. The dExo can effectively modulate various peripheral nerve-related cellular functions and is promising for cell-free biological therapeutics to enhance neural regeneration. Exosomes were isolated from hADMSCs with and without differentiation towards SC phenotype (i.e., dExo vs uExo). hADMSC-SCs secreted more growth factors compared to hADMSCs without differentiation. The dExo protected rat SCs from oxidative stress and enhanced endothelial cell migration and angiogenesis. dExo promoted axonal growth of sensory neurons differentiated from hiPSCs. miRNA sequencing analysis unveiled and compared the exosomal and cellular miRNA profiles.
Collapse
|
13
|
Augmenting Peripheral Nerve Regeneration with Adipose-Derived Stem Cells. Stem Cell Rev Rep 2022; 18:544-558. [PMID: 34417730 PMCID: PMC8858329 DOI: 10.1007/s12015-021-10236-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 02/03/2023]
Abstract
Peripheral nerve injuries (PNIs) are common and debilitating, cause significant health care costs for society, and rely predominately on autografts, which necessitate grafting a nerve section non-locally to repair the nerve injury. One possible approach to improving treatment is bolstering endogenous regenerative mechanisms or bioengineering new nervous tissue in the peripheral nervous system. In this review, we discuss critical-sized nerve gaps and nerve regeneration in rats, and summarize the roles of adipose-derived stem cells (ADSCs) in the treatment of PNIs. Several regenerative treatment modalities for PNI are described: ADSCs differentiating into Schwann cells (SCs), ADSCs secreting growth factors to promote peripheral nerve growth, ADSCs promoting myelination growth, and ADSCs treatments with scaffolds. ADSCs' roles in regenerative treatment and features are compared to mesenchymal stem cells, and the administration routes, cell dosages, and cell fates are discussed. ADSCs secrete neurotrophic factors and exosomes and can differentiate into Schwann cell-like cells (SCLCs) that share features with naturally occurring SCs, including the ability to promote nerve regeneration in the PNS. Future clinical applications are also discussed.
Collapse
|
14
|
Wang W, Gu MF, Wang ZF, Shen XM, Zhang J, Yang L. Let-7a-5p regulated by lncRNA-MEG3 promotes functional differentiation to Schwann cells from adipose derived stem cells via directly inhibiting RBPJ-mediating Notch pathway. Apoptosis 2021; 26:548-560. [PMID: 34409556 DOI: 10.1007/s10495-021-01685-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2021] [Indexed: 12/20/2022]
Abstract
Schwann cells (SCs) have important roles in supporting and repairing peripheral neurons, and thus have great potential for nerve injury treatment. Adipose tissue-derived stem cells (ADSCs) can be reliably induced to differentiate into SCs. However, the underlying molecular mechanisms are unclear. We explored the roles of MEG3/let-7a-5p/RBPJ axis in the differentiation into SCs from ADSCs. Primary ADSCs were induced to differentiate into SCs by appropriate reagents. ELISA, immunostaining, Western blotting, and qRT-PCR were employed to examine levels of SC-markers such as S100, GFAP, SOX10, p75NTR, GAP43, MPZ, β-NGF, BDNF, and NCAM and let-7 family, MEG3, RBPJ, and Notch signaling related proteins. Dual luciferase assay and RNA immunoprecipitation were performed to validate interactions of let-7a-5p/RBPJ mRNA and MEG3/let-7a-5p. Cultured ADSCs could be induced to differentiate into functional SCs. Let-7a-5p and let-7d-5p were elevated during the differentiation while MEG3 and RBPJ/Notch-signaling were suppressed. Let-7a-5p mimics promoted ADSC differentiation into SCs and up-regulated the levels of SC-related markers including S100, GFAP, SOX10, p75NTR, GAP43, MPZ, β-NGF, and NCAM, while RBPJ or MEG3 overexpression retarded the differentiation and reduced those levels. Let-7a-5p directly targeted RBPJ and MEG3 disinhibited Notch-RBPJ signaling via sponging let-7a-5p. RBPJ overexpression reversed the acceleration of let-7a-5p mimics on SC differentiation while let-7a-5p mimics blocked MEG3-mediated suppression on SC differentiation. Let-7a-5p sponged by MEG3 promotes differentiation of ADSCs into SCs via suppressing Notch signaling by targeting RBPJ. These findings shed light on mechanisms underlying the differentiation of ADSCs to SCs and provide avenues to accelerate the process.
Collapse
Affiliation(s)
- Wei Wang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Mei-Feng Gu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Zhi-Fei Wang
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410078, Hunan, China
| | - Xiang-Min Shen
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jie Zhang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Liang Yang
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410078, Hunan, China.
| |
Collapse
|
15
|
Interactive regulation of laryngeal cancer and neuroscience. Biochim Biophys Acta Rev Cancer 2021; 1876:188580. [PMID: 34129916 DOI: 10.1016/j.bbcan.2021.188580] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 06/10/2021] [Accepted: 06/10/2021] [Indexed: 01/11/2023]
Abstract
Nerve fibres are distributed throughout the body along with blood and lymphatic vessels. The intrinsic morphological characteristics of nerves and the general characteristics of secretions in the tumour microenvironment provide a solid theoretical basis for exploring how neuronal tissue can influence the progression of laryngeal cancer (LC). The central nervous system (CNS) and the peripheral nervous system (PNS) jointly control many aspects of cancer and have attracted widespread attention in the study of the progression, invasion and metastasis of tumour tissue banks. Stress activates the neuroendocrine response of the human hypothalamus-pituitary-adrenal (HPA) axis. LC cells induce nerve growth in the microenvironment by releasing neurotrophic factors (NTFs), and they can also stimulate neurite formation by secreting axons and axon guides. Conversely, nerve endings secrete factors that attract LC cells; this is known as perineural invasion (PNI) and promotes the progression of the associated cancer. In this paper, we summarize the systematic understanding of the role of neuroregulation in the LC tumour microenvironment (TME) and ways in which the TME accelerates nerve growth, which is closely related to the occurrence of LC.
Collapse
|
16
|
Zhang M, Wu JS, Xian HC, Chen BJ, Wang HF, Yu XH, Pang X, Dai L, Jiang J, Liang XH, Tang YL. CXCR5 induces perineural invasion of salivary adenoid cystic carcinoma by inhibiting microRNA-187. Aging (Albany NY) 2021; 13:15384-15399. [PMID: 34114971 PMCID: PMC8221347 DOI: 10.18632/aging.203097] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/11/2021] [Indexed: 02/05/2023]
Abstract
CXCR5 played critical roles in tumorigenesis and metastasis. Nevertheless, little was known about the involvement of CXCR5 in perineural invasion (PNI) of salivary adenoid cystic carcinoma (SACC). Here, we confirmed upregulation of CXCR5 in SACC specimens and cells and identified that CXCR5 exhibited a significant positive correlation with PNI. Functionally, knockdown of CXCR5 suppressed SACC cells migration, invasion and PNI ability, whereas CXCR5 overexpression displayed the opposite effects. Moreover, CXCR5 downregulated microRNA (miR)-187, which could competitively sponge S100A4. The PNI-inhibitory effect of CXCR5 knockdown or miR-187 overexpression could be reversed by elevated expression of S100A4. Conjointly, our data revealed that CXCR5 facilitated PNI through downregulating miR-187 to disinhibit S100A4 expression in SACC.
Collapse
Affiliation(s)
- Mei Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu 610041, China
| | - Jia-Shun Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu 610041, China
| | - Hong-Chun Xian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu 610041, China
| | - Bing-Jun Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu 610041, China
| | - Hao-Fan Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu 610041, China
| | - Xiang-Hua Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu 610041, China
| | - Xin Pang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu 610041, China
| | - Li Dai
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu 610041, China
| | - Jian Jiang
- Department of Head and Neck Surgery, Sichuan Cancer Hospital, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu 610041, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu 610041, China
| |
Collapse
|
17
|
Li K, Li X, Shi G, Lei X, Huang Y, Bai L, Qin C. Effectiveness and mechanisms of adipose-derived stem cell therapy in animal models of Parkinson's disease: a systematic review and meta-analysis. Transl Neurodegener 2021; 10:14. [PMID: 33926570 PMCID: PMC8081767 DOI: 10.1186/s40035-021-00238-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
Animal models provide an opportunity to assess the optimal treatment way and the underlying mechanisms of direct clinical application of adipose-derived stem cells (ADSCs). Previous studies have evaluated the effects of primitive and induced ADSCs in animal models of Parkinson's disease (PD). Here, eight databases were systematically searched for studies on the effects and in vivo changes caused by ADSC intervention. Quality assessment was conducted using a 10-item risk of bias tool. For the subsequent meta-analysis, study characteristics were extracted and effect sizes were computed. Ten out of 2324 published articles (n = 169 animals) were selected for further meta-analysis. After ADSC therapy, the rotation behavior (10 experiments, n = 156 animals) and rotarod performance (3 experiments, n = 54 animals) were improved (P < 0.000 01 and P = 0.000 3, respectively). The rotation behavior test reflected functional recovery, which may be due to the neurogenesis from neuronally differentiated ADSCs, resulting in a higher pooled effect size of standard mean difference (SMD) (- 2.59; 95% CI, - 3.57 to - 1.61) when compared to that of primitive cells (- 2.18; 95% CI, - 3.29 to - 1.07). Stratified analyses by different time intervals indicated that ADSC intervention exhibited a long-term effect. Following the transplantation of ADSCs, tyrosine hydroxylase-positive neurons recovered in the lesion area with pooled SMD of 13.36 [6.85, 19.86]. Transplantation of ADSCs is a therapeutic option that shows long-lasting effects in animal models of PD. The potential mechanisms of ADSCs involve neurogenesis and neuroprotective effects. The standardized induction of neural form of transplanted ADSCs can lead to a future application in clinical practice.
Collapse
Affiliation(s)
- Keya Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Xinyue Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Guiying Shi
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Xuepei Lei
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Yiying Huang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Lin Bai
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China.
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
18
|
Schwann-like cell conditioned medium promotes angiogenesis and nerve regeneration. Cell Tissue Bank 2021; 23:101-118. [PMID: 33837877 DOI: 10.1007/s10561-021-09920-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 03/27/2021] [Indexed: 12/29/2022]
Abstract
Vascular network reconstruction plays a pivotal role in the axonal regeneration and nerve function recovery after peripheral nerve injury. Increasing evidence indicates that Schwann cells (SCs) can promote nerve function repair, and the beneficial effects attributed to SCs therapy may exert their therapeutic effects through paracrine mechanisms. Recently, the previous research of our group demonstrated the promising neuroregenerative capacity of Schwann-like cells (SCLCs) derived from differentiated human embryonic stem cell-derived neural stem cells (hESC-NSCs) in vitro. Herein, the effects of SC-like cell conditioned medium (SCLC-CM) on angiogenesis and nerve regeneration were further explored. The assays were performed to show the pro-angiogenic effects of SCLC-CM, such as promoted endothelial cell proliferation, migration and tube formation in vitro. In addition, Sprague-Dawley rats were treated with SCLC-CM after sciatic nerve crush injury, SCLC-CM was conducive for the recovery of sciatic nerve function, which was mainly manifested in the SFI increase, the wet weight ratio of gastrocnemius muscle, as well as the number and thickness of myelin. The SCLC-CM treatment reduced the Evans blue leakage and increased the expression of CD34 microvessels. Furthermore, SCLC-CM upregulated the expressions of p-Akt and p-mTOR in endothelial cells. In conclusion, SCLC-CM promotes angiogenesis and nerve regeneration, it is expected to become a new treatment strategy for peripheral nerve injury.
Collapse
|
19
|
Li A, Pereira C, Hill EE, Vukcevich O, Wang A. In vitro, In vivo and Ex vivo Models for Peripheral Nerve Injury and Regeneration. Curr Neuropharmacol 2021; 20:344-361. [PMID: 33827409 PMCID: PMC9413794 DOI: 10.2174/1570159x19666210407155543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 01/29/2021] [Accepted: 03/29/2021] [Indexed: 11/22/2022] Open
Abstract
Peripheral Nerve Injuries (PNI) frequently occur secondary to traumatic injuries. Recovery from these injuries can be expectedly poor, especially in proximal injuries. In order to study and improve peripheral nerve regeneration, scientists rely on peripheral nerve models to identify and test therapeutic interventions. In this review, we discuss the best described and most commonly used peripheral nerve models that scientists have and continue to use to study peripheral nerve physiology and function.
Collapse
Affiliation(s)
- Andrew Li
- University of California Davis Ringgold standard institution - Hand and Upper Extremity Surgery, Division of Plastic Surgery, Department of Surgery Sacramento, California. United States
| | - Clifford Pereira
- University of California Davis Ringgold standard institution - Hand and Upper Extremity Surgery, Division of Plastic Surgery, Department of Surgery Sacramento, California. United States
| | - Elise Eleanor Hill
- University of California Davis Ringgold standard institution - Department of Surgery Sacramento, California. United States
| | - Olivia Vukcevich
- University of California Davis Ringgold standard institution - Surgery & Biomedical Engineering Sacramento, California. United States
| | - Aijun Wang
- University of California Davis - Surgery & Biomedical Engineering 4625 2nd Ave., Suite 3005 Sacramento Sacramento California 95817. United States
| |
Collapse
|
20
|
Huang Z, Powell R, Phillips JB, Haastert-Talini K. Perspective on Schwann Cells Derived from Induced Pluripotent Stem Cells in Peripheral Nerve Tissue Engineering. Cells 2020; 9:E2497. [PMID: 33213068 PMCID: PMC7698557 DOI: 10.3390/cells9112497] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Schwann cells play a crucial role in successful peripheral nerve repair and regeneration by supporting both axonal growth and myelination. Schwann cells are therefore a feasible option for cell therapy treatment of peripheral nerve injury. However, sourcing human Schwann cells at quantities required for development beyond research is challenging. Due to their availability, rapid in vitro expansion, survival, and integration within the host tissue, stem cells have attracted considerable attention as candidate cell therapies. Among them, induced pluripotent stem cells (iPSCs) with the associated prospects for personalized treatment are a promising therapy to take the leap from bench to bedside. In this critical review, we firstly focus on the current knowledge of the Schwann cell phenotype in regard to peripheral nerve injury, including crosstalk with the immune system during peripheral nerve regeneration. Then, we review iPSC to Schwann cell derivation protocols and the results from recent in vitro and in vivo studies. We finally conclude with some prospects for the use of iPSCs in clinical settings.
Collapse
Affiliation(s)
- Zhong Huang
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30623 Hannover, Germany;
- Center for Systems Neuroscience (ZSN) Hannover, 30559 Hannover, Germany
| | - Rebecca Powell
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK;
- UCL Centre for Nerve Engineering, University College London, London WC1E 6BT, UK
| | - James B. Phillips
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK;
- UCL Centre for Nerve Engineering, University College London, London WC1E 6BT, UK
| | - Kirsten Haastert-Talini
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30623 Hannover, Germany;
- Center for Systems Neuroscience (ZSN) Hannover, 30559 Hannover, Germany
| |
Collapse
|
21
|
Rhode SC, Beier JP, Ruhl T. Adipose tissue stem cells in peripheral nerve regeneration-In vitro and in vivo. J Neurosci Res 2020; 99:545-560. [PMID: 33070351 DOI: 10.1002/jnr.24738] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/16/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
After peripheral nerve injury, Schwann cells (SCs) are crucially involved in several steps of the subsequent regenerative processes, such as the Wallerian degeneration. They promote lysis and phagocytosis of myelin, secrete numbers of neurotrophic factors and cytokines, and recruit macrophages for a biological debridement. However, nerve injuries with a defect size of >1 cm do not show proper tissue regeneration and require a surgical nerve gap reconstruction. To find a sufficient alternative to the current gold standard-the autologous nerve transplant-several cell-based therapies have been developed and were experimentally investigated. One approach aims on the use of adipose tissue stem cells (ASCs). These are multipotent mesenchymal stromal cells that can differentiate into multiple phenotypes along the mesodermal lineage, such as osteoblasts, chondrocytes, and myocytes. Furthermore, ASCs also possess neurotrophic features, that is, they secrete neurotrophic factors like the nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3, ciliary neurotrophic factor, glial cell-derived neurotrophic factor, and artemin. They can also differentiate into the so-called Schwann cell-like cells (SCLCs). These cells share features with naturally occurring SCs, as they also promote nerve regeneration in the periphery. This review gives a comprehensive overview of the use of ASCs in peripheral nerve regeneration and peripheral nerve tissue engineering both in vitro and in vivo. While the sustainability of differentiation of ASCs to SCLCs in vivo is still questionable, ASCs used with different nerve conduits, such as hydrogels or silk fibers, have been shown to promote nerve regeneration.
Collapse
Affiliation(s)
- Sophie Charlotte Rhode
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Justus Patrick Beier
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Tim Ruhl
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
22
|
Ravenkamp M, Tchoukalova YD, Myers CE, Madsen CS, Shah MK, Zhang N, Lal D, Lott DG. The neurotrophic potential of human platelet lysate substitution for fetal bovine serum in glial induction culture medium. Neurosci Lett 2020; 730:135025. [PMID: 32387720 DOI: 10.1016/j.neulet.2020.135025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 01/23/2023]
Affiliation(s)
- Maile Ravenkamp
- Head and Neck Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Phoenix, AZ 85054, USA.
| | - Yourka D Tchoukalova
- Head and Neck Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Phoenix, AZ 85054, USA.
| | - Cheryl E Myers
- Head and Neck Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Phoenix, AZ 85054, USA.
| | - Cathy S Madsen
- Head and Neck Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Phoenix, AZ 85054, USA.
| | - Manisha K Shah
- Head and Neck Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Phoenix, AZ 85054, USA.
| | - Nan Zhang
- Department of Health Science Research, Section of Biostatistics, Mayo Clinic, Scottsdale, AZ 85059, USA.
| | - Devyani Lal
- Department of Otolaryngology - Head and Neck Surgery, Mayo Clinic, Phoenix, AZ 85054, USA.
| | - David G Lott
- Head and Neck Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; Department of Otolaryngology - Head and Neck Surgery, Mayo Clinic, Phoenix, AZ 85054, USA.
| |
Collapse
|
23
|
Adipose-derived mesenchymal stem cells protect against CMS-induced depression-like behaviors in mice via regulating the Nrf2/HO-1 and TLR4/NF-κB signaling pathways. Acta Pharmacol Sin 2020; 41:612-619. [PMID: 31796867 PMCID: PMC7468309 DOI: 10.1038/s41401-019-0317-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/09/2019] [Indexed: 12/27/2022] Open
Abstract
Increasing studies show that inflammatory processes may be involved in depressive disorders. Nuclear factor erythroid-2 related factor 2 (Nrf2) modulates tissue microglial M1 phenotypic changes to the M2 phenotype, which is implicated in protection against inflammatory diseases. We have reported that the adipose-derived mesenchymal stem cells (ADSCs) display anti-inflammatory activity. In this study we explored whether the mechanism of anti-inflammatory activity of ADSCs was related to Nrf2. ADSCs were isolated from mouse fat pads and intravenously administered to chronic mild stress (CMS)-exposed C57BL/6 mice at the dose of 1 × 106 once a week for 3 weeks. We showed that ADSC administration significantly remedied CMS-induced depressive-like behaviors in sucrose preference test, tail suspension test, and forced swim test accompanied by suppressing microglial activation and the expression of inflammatory factors including MCP-1, TNF-α, IL-1β, and IL-6. Furthermore, ADSC administration promoted both the expression of BDNF and TrkB, and promoted Nrf2/HO-1 signaling but suppressed TLR4/NF-κB signaling in brain tissue. In order to elucidate the role of Nrf2/HO-1 signaling in ADSC-caused neuroprotection, Nrf2-modified ADSCs were cocultured with BV2 microglial cells, then exposed to lipopolysaccharide (LPS). Downregulation of Nrf2 in ADSCs decreased the protective effects of ADSCs against LPS-induced microglial activation and M1 polarization. Nrf2 overexpression in ADSCs markedly suppressed LPS-induced TLR4 and NF-κB expression in microglial cells. These results suggest a possible antidepressive mechanism correlated with microglial polarization for anti-inflammatory agents, which may provide a new microglia-targeted strategy for depression therapy.
Collapse
|
24
|
Yi S, Zhang Y, Gu X, Huang L, Zhang K, Qian T, Gu X. Application of stem cells in peripheral nerve regeneration. BURNS & TRAUMA 2020; 8:tkaa002. [PMID: 32346538 PMCID: PMC7175760 DOI: 10.1093/burnst/tkaa002] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 02/07/2023]
Abstract
Traumatic peripheral nerve injury is a worldwide clinical issue with high morbidity. The severity of peripheral nerve injury can be classified as neurapraxia, axonotmesis or neurotmesis, according to Seddon's classification, or five different degrees according to Sunderland's classification. Patients with neurotmesis suffer from a complete transection of peripheral nerve stumps and are often in need of surgical repair of nerve defects. The applications of autologous nerve grafts as the golden standard for peripheral nerve transplantation meet some difficulties, including donor nerve sacrifice and nerve mismatch. Attempts have been made to construct tissue-engineered nerve grafts as supplements or even substitutes for autologous nerve grafts to bridge peripheral nerve defects. The incorporation of stem cells as seed cells into the biomaterial-based scaffolds increases the effectiveness of tissue-engineered nerve grafts and largely boosts the regenerative process. Numerous stem cells, including embryonic stem cells, neural stem cells, bone marrow mesenchymal stem cells, adipose stem cells, skin-derived precursor stem cells and induced pluripotent stem cells, have been used in neural tissue engineering. In the current review, recent trials of stem cell-based tissue-engineered nerve grafts have been summarized; potential concerns and perspectives of stem cell therapeutics have also been contemplated.
Collapse
Affiliation(s)
- Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Yu Zhang
- Nuclear Medicine Department, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiaokun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Li Huang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Kairong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Tianmei Qian
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
25
|
Moayeri A, Darvishi M, Amraei M. Homing of Super Paramagnetic Iron Oxide Nanoparticles (SPIONs) Labeled Adipose-Derived Stem Cells by Magnetic Attraction in a Rat Model of Parkinson's Disease. Int J Nanomedicine 2020; 15:1297-1308. [PMID: 32161459 PMCID: PMC7049746 DOI: 10.2147/ijn.s238266] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/07/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction Stem cell therapies for neurodegenerative diseases such as Parkinson’s disease (PD) are intended to replace lost dopaminergic neurons. The basis of this treatment is to guide the migration of transplanted cells into the target tissue or injury site. The aim of this study is an evaluation of the homing of superparamagnetic iron oxide nanoparticles (SPIONs) labeled adipose-derived stem cells (ADSC) by an external magnetic field in a rat model of PD. Methods ADSCs were obtained from perinephric regions of male adult rats and cultured in a DMEM medium. ADSC markers were assessed by immunostaining with CD90, CD105, CD49d, and CD45. The SPION was coated using poly-L-lysine hydrobromide and transfection was determined in rat ADSC using the GFP reporter gene. For this in vivo study, rats with PD were divided into five groups: a positive control group, a control group with PD (lesion with 6-HD injection), and three treatment groups: the PD/ADSC group (PD transplant with ADSCs transfected by BrdU), PD/ADSC/SPION group (PD transplant with ADSCs labeled with SPION and transfected by GFP), and the PD/ADSC/SPION/EM group (PD transplant with ADSCs labeled with SPION and transfected by GFP induced with external magnet). Results ADSCs were immunoreactive to fat markers CD90 (90.73±1.7), CD105 (87.4±2.9) and CD49d (79.6±2.6), with negative immunostaining at the hematopoietic stem cell marker (CD45: 1.4±0.4). The efficiency of cells with SPION/PLL was about 96% of ADSC. The highest number of GFP-positive cells was in the ADSC/SPION/EM group (54.5±1.3), which was significantly different from that in ADSC/SPION group (30.83±3 and P<0.01). Conclusion Transfection of ADSC by SPION/PLL is an appropriate protocol for cell therapy. External magnets can be used for the delivery and homing of transplanted stem cells in the target tissue.
Collapse
Affiliation(s)
- Ardeshir Moayeri
- Department of Anatomy, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Marzieh Darvishi
- Department of Anatomy, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran.,Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Mansour Amraei
- Department of Physiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
26
|
Effect of Induction Time on the Proliferation and Differentiation of Induced Schwann-Like Cells from Adipose-Derived Stem Cells. Cell Mol Neurobiol 2020; 40:1105-1116. [DOI: 10.1007/s10571-020-00795-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 01/18/2020] [Indexed: 12/15/2022]
|
27
|
The Cellular and Molecular Patterns Involved in the Neural Differentiation of Adipose-Derived Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1298:23-41. [PMID: 32514816 DOI: 10.1007/5584_2020_547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Injuries to the nervous system cause serious problems among affected patients by preventing them from the possibility of living a normal life. As this tissue possesses a reduced capacity of self-regeneration currently, lots of different strategies are being developed in order to make the regeneration in the nervous system possible. Among them, tissue engineering and stem cell-based therapies are to date very exploded fields and tremendous progress has been made in this direction. As the two main components of the nervous system, react differently to injuries and behave different during disease, it is clear that two separate regeneration approaches have been taken into consideration during development of treatment. Special attention is constantly given to the potential of adipose-derived stem cells for this kind of application. Due to the fact that they present remarkable properties, they can easily be obtained and have demonstrated that are capable of engaging in neural and glial lineages, adipose-derived stem cells are promising tools for the field of nervous system regeneration. Moreover, new insights into epigenetic control and modifications during the differentiation of adipose-derived stem cells towards the neural liege could provide new methods to maximize the regeneration process. In this review, we summarize the current applications of adipose-derived stem cells for neural regeneration and discuss in-depth molecular patterns involved in the differentiation of adipose-derived stem cells in neuron-like cells and Schwann-like cells.
Collapse
|
28
|
Ge P, Guo Y, Shen J. IcarisideII facilitates the differentiation of ADSCs to SCs via let-7i/STAT3 axis to preserve erectile function. Biol Res 2019; 52:54. [PMID: 31581950 PMCID: PMC6777035 DOI: 10.1186/s40659-019-0262-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND IcarisideII (ICAII) could promote the differentiation of adipose tissue-derived stem cells (ADSCs) to Schwann cells (SCs), leading to improvement of erectile function (EF) and providing a realistic therapeutic option for the treatment of erectile dysfunction (ED). However, the underlying molecular mechanisms of ADSCs and ICAII in this process remain largely unclear. METHODS ADSCs were treated with different concentrations of ICAII. Cell proliferation was determined by MTT assay. qRT-PCR and western blot were performed to detect expressions of SCs markers, signal transducer and activator of transcription-3 (STAT3), and microRNA-let-7i (let-7i). Luciferase reporter assay was conducted to verify the regulatory relationship between let-7i and STAT3. The detection of intracavernosal pressure (ICP) and the ratio of ICP/mean arterial pressure (MAP) were used to evaluate the EF in bilateral cavernous nerve injury (BCNI) rat models. RESULTS ICAII promoted cell proliferation of ADSCs in a dose-dependent manner. The mRNA and protein levels of SCs markers were increased by ICAII treatment in a dose-dependent manner in ADSCs. Moreover, let-7i was significantly decreased in ICAII-treated ADSCs and upregulation of let-7i attenuated ICAII-induced promotion of SCs markers. In addition, STAT3 was a direct target of let-7i and upregulated in ICAII-treated ADSCs. Interestingly, overexpression of STAT3 abated the let-7i-mediated inhibition effect on differentiation of ADSCs to SCs and rescued the ICAII-mediated promotion effect on it. Besides, combination treatment of ADSCs and ICAII preserved the EF of BCNI rat models, which was undermined by let-7i overexpression. CONCLUSION ICAII was effective for preserving EF by promoting the differentiation of ADSCs to SCs via modulating let-7i/STAT3 pathway.
Collapse
Affiliation(s)
- Pingyu Ge
- Department of Urology Surgery, the First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71, Baoshan North Road, Guiyang, 550001, Guizhou, China.
| | - Yinxue Guo
- Department of Nephrology, the First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Jun Shen
- Department of Urology Surgery, the First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71, Baoshan North Road, Guiyang, 550001, Guizhou, China
| |
Collapse
|
29
|
Gao S, Guo X, Zhao S, Jin Y, Zhou F, Yuan P, Cao L, Wang J, Qiu Y, Sun C, Kang Z, Gao F, Xu W, Hu X, Yang D, Qin Y, Ning K, Shaw PJ, Zhong G, Cheng L, Zhu H, Gao Z, Chen X, Xu J. Differentiation of human adipose-derived stem cells into neuron/motoneuron-like cells for cell replacement therapy of spinal cord injury. Cell Death Dis 2019; 10:597. [PMID: 31395857 PMCID: PMC6687731 DOI: 10.1038/s41419-019-1772-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/24/2019] [Accepted: 05/31/2019] [Indexed: 01/06/2023]
Abstract
Human adipose-derived stem cells (hADSCs) are increasingly presumed to be a prospective stem cell source for cell replacement therapy in various degenerative and/or traumatic diseases. The potential of trans-differentiating hADSCs into motor neuron cells indisputably provides an alternative way for spinal cord injury (SCI) treatment. In the present study, a stepwise and efficient hADSC trans-differentiation protocol with retinoic acid (RA), sonic hedgehog (SHH), and neurotrophic factors were developed. With this protocol hADSCs could be converted into electrophysiologically active motoneuron-like cells (hADSC-MNs), which expressed both a cohort of pan neuronal markers and motor neuron specific markers. Moreover, after being primed for neuronal differentiation with RA/SHH, hADSCs were transplanted into SCI mouse model and they survived, migrated, and integrated into injured site and led to partial functional recovery of SCI mice. When ablating the transplanted hADSC-MNs harboring HSV-TK-mCherry overexpression system with antivirial Ganciclovir (GCV), functional relapse was detected by motor-evoked potential (MEP) and BMS assays, implying that transplanted hADSC-MNs participated in rebuilding the neural circuits, which was further confirmed by retrograde neuronal tracing system (WGA). GFP-labeled hADSC-MNs were subjected to whole-cell patch-clamp recording in acute spinal cord slice preparation and both action potentials and synaptic activities were recorded, which further confirmed that those pre-conditioned hADSCs indeed became functionally active neurons in vivo. As well, transplanted hADSC-MNs largely prevented the formation of injury-induced cavities and exerted obvious immune-suppression effect as revealed by preventing astrocyte reactivation and favoring the secretion of a spectrum of anti-inflammatory cytokines and chemokines. Our work suggests that hADSCs can be readily transformed into MNs in vitro, and stay viable in spinal cord of the SCI mouse and exert multi-therapeutic effects by rebuilding the broken circuitry and optimizing the microenvironment through immunosuppression.
Collapse
Affiliation(s)
- Shane Gao
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xuanxuan Guo
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Simeng Zhao
- iHuman Institute, Shanghai Science and Technology University, Shanghai, 201210, China
| | - Yinpeng Jin
- Shanghai Public Health Clinical Center, Fudan University, JinShan, Shanghai, 201508, China
| | - Fei Zhou
- Department of Neurology, Third Affiliated Hospital of Navy Military Medical University, Shanghai, 200438, China
| | - Ping Yuan
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China
| | - Limei Cao
- Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China
| | - Jian Wang
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yue Qiu
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Chenxi Sun
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhanrong Kang
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 200137, China
| | - Fengjuan Gao
- Zhoupu hospital, Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Wei Xu
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xiao Hu
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China
| | - Danjing Yang
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Ying Qin
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Ke Ning
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ, UK
| | - Pamela J Shaw
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ, UK
| | - Guisheng Zhong
- iHuman Institute, Shanghai Science and Technology University, Shanghai, 201210, China.
| | - Liming Cheng
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Hongwen Zhu
- Tianjin Hospital, Tianjin, 300211, China. .,BOE Technology Group Co., Ltd., Beijing, 100176, China.
| | - Zhengliang Gao
- Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Xu Chen
- Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China.
| | - Jun Xu
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
30
|
Yang H, Hao D, Liu C, Huang D, Chen B, Fan H, Liu C, Zhang L, Zhang Q, An J, Zhao J. Generation of functional dopaminergic neurons from human spermatogonial stem cells to rescue parkinsonian phenotypes. Stem Cell Res Ther 2019; 10:195. [PMID: 31248447 PMCID: PMC6598262 DOI: 10.1186/s13287-019-1294-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 01/08/2023] Open
Abstract
Background Recent progress in the induced generation of dopaminergic (DA) neurons from different types of stem cells or reprogrammed somatic cells holds tremendous potential for the treatment of Parkinson’s disease (PD). However, the lack of a reliable source for cell replacement therapy remains a major limitation in the treatment of human neurological disorders. Additionally, the current protocols for in vitro differentiation or cell reprogramming to generate human DA neurons are laborious, time-consuming, and expensive, and efficient conversion of human spermatogonial stem cells (hSSCs) to functional DA neurons has not yet been achieved. Methods Primary hSSCs from testicular tissues of patients were exposed to an improved induction system, which consisted mainly of olfactory ensheathing cell conditioned culture medium (OECCM) and a set of defined cell-extrinsic factors and small molecules. Morphological changes were assessed, along with the expression of various DA neuron phenotypic markers (e.g., Tuj-1, TH, Nurr1, DAT) and several critical pro-DA neurogenesis effectors (e.g., EN-1, Pitx3, Foxa2, Lmx1a, Lmx1b, and OTX2). In addition, transcriptome analysis was used to further evaluate the genetic similarity between the artificially differentiated DA neurons and genuine ones. Concomitantly, the functional properties of converted DA neurons including synapse formation, dopamine release, electrophysiological activity, and neuron-specific Ca2+ signaling images were determined. Finally, hSSCs in the early stage of induction were evaluated for survival, differentiation, migration, tumorigenicity in the mouse striatum, and improvement of functional deficits in MPTP-induced PD animals. Results The hSSC-derived neurons not only acquired neuronal morphological features but also expressed various phenotypic genes and protein characteristic of DA neurons and several effectors critical for pro-DA neurogenesis. Strikingly, as the period of induction was prolonged, expression of the critical molecules for DA neuron epigenetic status gradually increased while hSSC-specific markers sharply decreased. After 3 weeks of induction, the transdifferentiation efficiency reached 21%. In addition, hierarchical clustering analysis showed that the differentiated DA neurons closely resembled genuine ones. Furthermore, the hSSC-derived neurons gained sophisticated functional properties of wild-type DA neurons, and pro-induced hSSCs efficiently survived, migrated, and differentiated into DA neurons without tumorigenesis after transplantation into mouse striatum, leading to improvement of functional deficits in PD animals. Conclusions The results showed that, using the present improved straightforward approach, hSSCs could acquire DA neuron morphological features and functional properties and rescue parkinsonian phenotypes. Our strategy for the conversion of hSSCs into DA neurons is very efficient and thus may provide an alternative approach suitable for clinical cell therapy to treat neurodegenerative diseases including PD.
Collapse
Affiliation(s)
- Hao Yang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China.
| | - Dingjun Hao
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China.,Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Cheng Liu
- Department of Foot and Ankle Surge, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Dageng Huang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Bo Chen
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Hong Fan
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Cuicui Liu
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Lingling Zhang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Qian Zhang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Jing An
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Jingjing Zhao
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| |
Collapse
|
31
|
Zhang M, Li Z, Wang H, Wang S, Yu X, Wu J, Pang X, Wu J, Yang X, Tang Y, Li L, Liang X, Zheng M, Tang Y. MIF promotes perineural invasion through EMT in salivary adenoid cystic carcinoma. Mol Carcinog 2019; 58:898-912. [PMID: 30667094 DOI: 10.1002/mc.22979] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/16/2019] [Accepted: 01/16/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Mei Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Zhu‐feng Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Hao‐fan Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Sha‐sha Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Xiang‐hua Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Jing‐biao Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Xin Pang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Jia‐shun Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Xiao Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Ya‐jie Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education)Hubei Provincial Cooperative Innovation Center of Industrial FermentationHubei Key Laboratory of Industrial MicrobiologyHubei University of TechnologyWuhanChina
| | - Li Li
- Department of StomatologyZhoushan HospitalWenzhou Medical UniversityZhoushanZhejiangChina
| | - Xin‐hua Liang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Min Zheng
- Department of StomatologyZhoushan HospitalWenzhou Medical UniversityZhoushanZhejiangChina
| | - Ya‐ling Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| |
Collapse
|
32
|
Ramli K, Aminath Gasim I, Ahmad AA, Hassan S, Law ZK, Tan GC, Baharuddin A, Naicker AS, Htwe O, Mohammed Haflah NH, B H Idrus R, Abdullah S, Ng MH. Human bone marrow-derived MSCs spontaneously express specific Schwann cell markers. Cell Biol Int 2019; 43:233-252. [PMID: 30362196 DOI: 10.1002/cbin.11067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/07/2018] [Indexed: 12/15/2022]
Abstract
In peripheral nerve injuries, Schwann cells (SC) play pivotal roles in regenerating damaged nerve. However, the use of SC in clinical cell-based therapy is hampered due to its limited availability. In this study, we aim to evaluate the effectiveness of using an established induction protocol for human bone marrow derived-MSC (hBM-MSCs) transdifferentiation into a SC lineage. A relatively homogenous culture of hBM-MSCs was first established after serial passaging (P3), with profiles conforming to the minimal criteria set by International Society for Cellular Therapy (ISCT). The cultures (n = 3) were then subjected to a series of induction media containing β-mercaptoethanol, retinoic acid, and growth factors. Quantitative RT-PCR, flow cytometry, and immunocytochemistry analyses were performed to quantify the expression of specific SC markers, that is, S100, GFAP, MPZ and p75 NGFR, in both undifferentiated and transdifferentiated hBM-MSCs. Based on these analyses, all markers were expressed in undifferentiated hBM-MSCs and MPZ expression (mRNA transcripts) was consistently detected before and after transdifferentiation across all samples. There was upregulation at the transcript level of more than twofolds for NGF, MPB, GDNF, p75 NGFR post-transdifferentiation. This study highlights the existence of spontaneous expression of specific SC markers in cultured hBM-MSCs, inter-donor variability and that MSC transdifferentiation is a heterogenous process. These findings strongly oppose the use of a single marker to indicate SC fate. The heterogenous nature of MSC may influence the efficiency of SC transdifferentiation protocols. Therefore, there is an urgent need to re-define the MSC subpopulations and revise the minimal criteria for MSC identification.
Collapse
Affiliation(s)
- Khairunnisa Ramli
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Ifasha Aminath Gasim
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Amir Adham Ahmad
- Department of Orthopaedics, School of Medicine, International Medical University, Negeri Sembilan, Malaysia
| | - Shariful Hassan
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Zhe Kang Law
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Azmi Baharuddin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Amaramalar Selvi Naicker
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ohnmar Htwe
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nor Hazla Mohammed Haflah
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ruszymah B H Idrus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shalimar Abdullah
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Min Hwei Ng
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
33
|
Lo Furno D, Mannino G, Pellitteri R, Zappalà A, Parenti R, Gili E, Vancheri C, Giuffrida R. Conditioned Media From Glial Cells Promote a Neural-Like Connexin Expression in Human Adipose-Derived Mesenchymal Stem Cells. Front Physiol 2018; 9:1742. [PMID: 30555356 PMCID: PMC6282092 DOI: 10.3389/fphys.2018.01742] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
The expression of neuronal and glial connexins (Cxs) has been evaluated in adipose-derived mesenchymal stem cells (ASCs) whose neural differentiation was promoted by a conditioned medium (CM) obtained from cultures of olfactory ensheathing cells (OECs) or Schwann cells (SCs). By immunocytochemistry and flow cytometer analysis it was found that Cx43 was already considerably expressed in naïve ASCs and further increased after 24 h and 7 days from CM exposition. Cx32 and Cx36 were significantly improved in conditioned cultures compared to control ASCs, whereas a decreased expression was noticed in the absence of CM treatments. Cx47 was virtually absent in any conditions. Altogether, high basal levels and induced increases of Cx43 expression suggest a potential attitude of ASCs toward an astrocyte differentiation, whereas the lack of Cx47 would indicate a poor propensity of ASCs to become oligodendrocytes. CM-evoked Cx32 and Cx36 increases showed that a neuronal- or a SC-like differentiation can be promoted by using this strategy. Results further confirm that environmental cues can favor an ASC neural differentiation, either as neuronal or glial elements. Of note, the use of glial products present in CM rather than the addition of chemical agents to achieve such differentiation would resemble "more physiological" conditions of differentiation. As a conclusion, the overexpression of typical neural Cxs would indicate the potential capability of neural-like ASCs to interact with neighboring neural cells and microenvironment.
Collapse
Affiliation(s)
- Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Giuliana Mannino
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Rosalia Pellitteri
- Institute of Neurological Sciences, National Research Council, Catania, Italy
| | - Agata Zappalà
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Elisa Gili
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Carlo Vancheri
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| |
Collapse
|
34
|
Uz M, Das SR, Ding S, Sakaguchi DS, Claussen JC, Mallapragada SK. Advances in Controlling Differentiation of Adult Stem Cells for Peripheral Nerve Regeneration. Adv Healthc Mater 2018; 7:e1701046. [PMID: 29656561 DOI: 10.1002/adhm.201701046] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 01/08/2018] [Indexed: 01/01/2023]
Abstract
Adult stems cells, possessing the ability to grow, migrate, proliferate, and transdifferentiate into various specific phenotypes, constitute a great asset for peripheral nerve regeneration. Adult stem cells' ability to undergo transdifferentiation is sensitive to various cell-to-cell interactions and external stimuli involving interactions with physical, mechanical, and chemical cues within their microenvironment. Various studies have employed different techniques for transdifferentiating adult stem cells from distinct sources into specific lineages (e.g., glial cells and neurons). These techniques include chemical and/or electrical induction as well as cell-to-cell interactions via co-culture along with the use of various 3D conduit/scaffold designs. Such scaffolds consist of unique materials that possess controllable physical/mechanical properties mimicking cells' natural extracellular matrix. However, current limitations regarding non-scalable transdifferentiation protocols, fate commitment of transdifferentiated stem cells, and conduit/scaffold design have required new strategies for effective stem cells transdifferentiation and implantation. In this progress report, a comprehensive review of recent advances in the transdifferentiation of adult stem cells via different approaches along with multifunctional conduit/scaffolds designs is presented for peripheral nerve regeneration. Potential cellular mechanisms and signaling pathways associated with differentiation are also included. The discussion with current challenges in the field and an outlook toward future research directions is concluded.
Collapse
Affiliation(s)
- Metin Uz
- Department of Chemical and Biological Engineering Iowa State University Ames IA 50011 USA
| | - Suprem R. Das
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
- Division of Materials Science and Engineering Ames Laboratory Ames IA 50011 USA
| | - Shaowei Ding
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
| | - Donald S. Sakaguchi
- Neuroscience Program Iowa State University Ames IA 50011 USA
- Department of Genetics Development and Cell Biology Iowa State University Ames IA 50011 USA
| | - Jonathan C. Claussen
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
- Division of Materials Science and Engineering Ames Laboratory Ames IA 50011 USA
| | - Surya K. Mallapragada
- Department of Chemical and Biological Engineering Iowa State University Ames IA 50011 USA
- Department of Genetics Development and Cell Biology Iowa State University Ames IA 50011 USA
| |
Collapse
|