1
|
Liu M, Lu Y, Sun F, Li Y, Wu J, Zou Q. The Nerve-Induced Adipose Stem Cells Promote Nerve Repair in Stress Urinary Incontinence by Regulating Schwann Cell Repair Phenotype Conversion Through Activation of the Notch Pathway. Mol Neurobiol 2025:10.1007/s12035-025-04704-z. [PMID: 39881114 DOI: 10.1007/s12035-025-04704-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
Stress urinary incontinence (SUI) currently lacks effective treatment options, and the restoration of neurological function remains a major challenge, with unmet clinical needs. Research has indicated that adipose-derived stem cells (ADSCs) can be induced to differentiate into neural-induced adipose-derived stem cells (NI-ADSCs) under specific inductive conditions, exhibiting excellent neuroregenerative capabilities. ADSCs were obtained from female SD rats and induced into NI-ADSCs. In vitro, NI-ADSCs were co-cultured with Schwann cells (SCs) to investigate their effects on SC proliferation and repair phenotype transition and further explore its underlying mechanism. In vivo, a rat model of SUI was established using a bilateral pudendal nerve transection method. NI-ADSCs were injected into the urethral sphincter to evaluate their effects on urodynamics, muscle angiogenesis, and neural repair in SUI rats, while also exploring the mechanisms of neural repair. This study used EGF, FGF, and B27 to induce ADSCs into NI-ADSCs expressing neural induction markers (MAP, Nestin, and PAX6). In vitro experiments found no significant difference in the proliferation of L6 and RSC96 between NI-ADSCs and ADSCs (p > 0.05). However, when co-cultured with NI-ADSCs, SCs showed upregulated expression of repair-related phenotypic markers (BDNF, GDNF, and GFAP). In this phenotypic transformation process, the expression of Notch-related pathway proteins (Notch1, NICD, and Hes1) was increased, and the use of DAPT (a Notch pathway inhibitor) could suppress the SC repair phenotype transformation. In vivo, experiments revealed that intraurethral injection of NI-ADSCs significantly promoted the expression of neural marker (S100β) and demyelination markers (GFAP) and urodynamic recovery in SUI rats, while DAPT inhibited its neural repair effect. In summary, our study demonstrates that NI-ADSCs can promote nerve regeneration by promoting and maintaining the repair-related phenotype of SCs. The underlying mechanism may be related to the activation of the Notch signaling pathway.
Collapse
Affiliation(s)
- Ming Liu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, No. 20 East Yuhuangding Road, Yantai, 264000, Shandong, China
- Second Clinical Medical College, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Youyi Lu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, No. 20 East Yuhuangding Road, Yantai, 264000, Shandong, China
| | - Fengze Sun
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, No. 20 East Yuhuangding Road, Yantai, 264000, Shandong, China
| | - Yongwei Li
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, No. 20 East Yuhuangding Road, Yantai, 264000, Shandong, China
| | - Jitao Wu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, No. 20 East Yuhuangding Road, Yantai, 264000, Shandong, China.
| | - Qingsong Zou
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, No. 20 East Yuhuangding Road, Yantai, 264000, Shandong, China.
| |
Collapse
|
2
|
El Masri J, Fadlallah H, Al Sabsabi R, Afyouni A, Al-Sayegh M, Abou-Kheir W. Adipose-Derived Stem Cell Therapy in Spinal Cord Injury. Cells 2024; 13:1505. [PMID: 39273075 PMCID: PMC11394073 DOI: 10.3390/cells13171505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Spinal cord injury (SCI) is a serious condition accompanied by severe adverse events that affect several aspects of the patient's life, such as motor, sensory, and functional impairment. Despite its severe consequences, definitive treatment for these injuries is still missing. Therefore, researchers have focused on developing treatment strategies aimed at ensuring full recovery post-SCI. Accordingly, attention has been drawn toward cellular therapy using mesenchymal stem cells. Considering their wide availability, decreased immunogenicity, wide expansion capacity, and impressive effectiveness in many therapeutic approaches, adipose-derived stem cell (ADSC) injections in SCI cases have been investigated and showed promising results. In this review, SCI pathophysiology and ADSC transplantation benefits are discussed independently, together with SCI animal models and adipose stem cell preparation and application techniques. The mechanisms of healing in an SCI post-ADSC injection, the outcomes of this therapeutic approach, and current clinical trials are also deliberated, in addition to the challenges and future perspectives, aiming to encourage further research in this field.
Collapse
Affiliation(s)
- Jad El Masri
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon; (J.E.M.); (H.F.)
- Faculty of Medical Sciences, Lebanese University, Beirut 1533, Lebanon; (R.A.S.); (A.A.)
| | - Hiba Fadlallah
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon; (J.E.M.); (H.F.)
| | - Rahaf Al Sabsabi
- Faculty of Medical Sciences, Lebanese University, Beirut 1533, Lebanon; (R.A.S.); (A.A.)
| | - Ahmad Afyouni
- Faculty of Medical Sciences, Lebanese University, Beirut 1533, Lebanon; (R.A.S.); (A.A.)
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, Abu Dhabi 2460, United Arab Emirates
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon; (J.E.M.); (H.F.)
| |
Collapse
|
3
|
Choi J, Gang S, Ramalingam M, Hwang J, Jeong H, Yoo J, Cho HH, Kim BC, Jang G, Jeong HS, Jang S. BML-281 promotes neuronal differentiation by modulating Wnt/Ca 2+ and Wnt/PCP signaling pathway. Mol Cell Biochem 2024; 479:2391-2403. [PMID: 37768498 DOI: 10.1007/s11010-023-04857-2] [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: 07/11/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023]
Abstract
Histone deacetylase (HDAC) inhibitors promote differentiation through post-translational modifications of histones. BML-281, an HDAC6 inhibitor, has been known to prevent tumors, acute dextran sodium sulfate-associated colitis, and lung injury. However, the neurogenic differentiation effect of BML-281 is poorly understood. In this study, we investigated the effect of BML-281 on neuroblastoma SH-SY5Y cell differentiation into mature neurons by immunocytochemistry (ICC), reverse transcriptase PCR (RT-PCR), quantitative PCR (qPCR), and western blotting analysis. We found that the cells treated with BML-281 showed neurite outgrowth and morphological changes into mature neurons under a microscope. It was confirmed that the gene expression of neuronal markers (NEFL, MAP2, Tuj1, NEFH, and NEFM) was increased with certain concentrations of BML-281. Similarly, the protein expression of neuronal markers (NeuN, Synaptophysin, Tuj1, and NFH) was upregulated with BML-281 compared to untreated cells. Following treatment with BML-281, the expression of Wnt5α increased, and downstream pathways were activated. Interestingly, both Wnt/Ca2+ and Wnt/PCP pathways activated and regulated PKC, Cdc42, RhoA, Rac1/2/3, and p-JNK. Therefore, BML-281 induces the differentiation of SH-SY5Y cells into mature neurons by activating the non-canonical Wnt signaling pathway. From these results, we concluded that BML-281 might be a novel drug to differentiation into neuronal cells through the regulation of Wnt signaling pathway to reduce the neuronal cell death.
Collapse
Affiliation(s)
- Jiyun Choi
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| | - Seoyeon Gang
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
- Department of Pre-Medical Science, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| | - Mahesh Ramalingam
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| | - Jinsu Hwang
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| | - Haewon Jeong
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| | - Jin Yoo
- Department of Physiological Education, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyong-Ho Cho
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Byeong C Kim
- Department of Neurology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Geupil Jang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea.
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea.
| |
Collapse
|
4
|
Gentile AM, Lhamyani S, Mengual-Mesa M, García-Fuentes E, Bermúdez-Silva FJ, Rojo-Martínez G, Clemente-Postigo M, Rodriguez-Cañete A, Olveira G, El Bekay R. MiR-221-3p/222-3p Cluster Expression in Human Adipose Tissue Is Related to Obesity and Type 2 Diabetes. Int J Mol Sci 2023; 24:17449. [PMID: 38139277 PMCID: PMC10744326 DOI: 10.3390/ijms242417449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The progression of obesity and type 2 diabetes (T2D) is intricately linked with adipose tissue (AT) angiogenesis. Despite an established network of microRNAs (miRNAs) regulating AT function, the specific role of angiogenic miRNAs remains less understood. The miR-221/222 cluster has recently emerged as being associated with antiangiogenic activity. However, no studies have explored its role in human AT amidst the concurrent development of obesity and T2D. Therefore, this study aims to investigate the association between the miR-221-3p/222-3p cluster in human AT and its regulatory network with obesity and T2D. MiR-221-3p/222-3p and their target gene (TG) expression levels were quantified through qPCR in visceral (VAT) and subcutaneous (SAT) AT from patients (n = 33) categorized based on BMI as normoweight (NW) and obese (OB) and by glycemic status as normoglycemic (NG) and type 2 diabetic (T2D) subjects. In silico analyses of miR-221-3p/222-3p and their TGs were conducted to identify pertinent signaling pathways. The results of a multivariate analysis, considering the simultaneous expression of miR-221-3p and miR-222-3p as dependent variables, revealed statistically significant distinctions when accounting for variables such as tissue depot, obesity, sex, and T2D as independent factors. Furthermore, both miRNAs and their TGs exhibited differential expression patterns based on obesity severity, glycemic status, sex, and type of AT depot. Our in silico analysis indicated that miR-221-3p/222-3p cluster TGs predominantly participate in angiogenesis, WNT signaling, and apoptosis pathways. In conclusion, these findings underscore a promising avenue for future research, emphasizing the miR-221-3p/222-3p cluster and its associated regulatory networks as potential targets for addressing obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Adriana-Mariel Gentile
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29580 Málaga, Spain; (A.-M.G.); (S.L.); (E.G.-F.); (F.-J.B.-S.); (G.R.-M.); (G.O.)
- Clinical Unit of Endocrinology and Nutrition, University Regional Hospital of Málaga, 29009 Málaga, Spain
| | - Said Lhamyani
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29580 Málaga, Spain; (A.-M.G.); (S.L.); (E.G.-F.); (F.-J.B.-S.); (G.R.-M.); (G.O.)
- Clinical Unit of Endocrinology and Nutrition, University Regional Hospital of Málaga, 29009 Málaga, Spain
- Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Mengual-Mesa
- Andalucía Tech, Faculty of Health Sciences, Department of Systems and Automation Engineering, School of Industrial Engineering, Universidad de Málaga, Teatinos Campus, 29071 Málaga, Spain;
| | - Eduardo García-Fuentes
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29580 Málaga, Spain; (A.-M.G.); (S.L.); (E.G.-F.); (F.-J.B.-S.); (G.R.-M.); (G.O.)
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Málaga, Spain
| | - Francisco-Javier Bermúdez-Silva
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29580 Málaga, Spain; (A.-M.G.); (S.L.); (E.G.-F.); (F.-J.B.-S.); (G.R.-M.); (G.O.)
- Clinical Unit of Endocrinology and Nutrition, University Regional Hospital of Málaga, 29009 Málaga, Spain
- The Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Gemma Rojo-Martínez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29580 Málaga, Spain; (A.-M.G.); (S.L.); (E.G.-F.); (F.-J.B.-S.); (G.R.-M.); (G.O.)
- Clinical Unit of Endocrinology and Nutrition, University Regional Hospital of Málaga, 29009 Málaga, Spain
- The Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mercedes Clemente-Postigo
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Institute of Biomedical Research in Málaga (IBIMA)-Bionand Platform, 29590 Málaga, Spain
- Department of Cell Biology, Genetics, and Physiology, Faculty of Science, University of Málaga, 29010 Málaga, Spain
| | - Alberto Rodriguez-Cañete
- Unidad de Gestión Clínica de Cirugía General, Digestiva y Trasplantes, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain;
| | - Gabriel Olveira
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29580 Málaga, Spain; (A.-M.G.); (S.L.); (E.G.-F.); (F.-J.B.-S.); (G.R.-M.); (G.O.)
- Clinical Unit of Endocrinology and Nutrition, University Regional Hospital of Málaga, 29009 Málaga, Spain
- The Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Medicina y Cirugía, Universidad de Málaga, 29010 Málaga, Spain
| | - Rajaa El Bekay
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29580 Málaga, Spain; (A.-M.G.); (S.L.); (E.G.-F.); (F.-J.B.-S.); (G.R.-M.); (G.O.)
- Clinical Unit of Endocrinology and Nutrition, University Regional Hospital of Málaga, 29009 Málaga, Spain
- Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- IBIMA-Plataforma Bionand, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| |
Collapse
|
5
|
Wu H, Fan Y, Zhang M. Advanced Progress in the Role of Adipose-Derived Mesenchymal Stromal/Stem Cells in the Application of Central Nervous System Disorders. Pharmaceutics 2023; 15:2637. [PMID: 38004615 PMCID: PMC10674952 DOI: 10.3390/pharmaceutics15112637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/29/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Currently, adipose-derived mesenchymal stromal/stem cells (ADMSCs) are recognized as a highly promising material for stem cell therapy due to their accessibility and safety. Given the frequently irreversible damage to neural cells associated with CNS disorders, ADMSC-related therapy, which primarily encompasses ADMSC transplantation and injection with exosomes derived from ADMSCs or secretome, has the capability to inhibit inflammatory response and neuronal apoptosis, promote neural regeneration, as well as modulate immune responses, holding potential as a comprehensive approach to treat CNS disorders and improve prognosis. Empirical evidence from both experiments and clinical trials convincingly demonstrates the satisfactory safety and efficacy of ADMSC-related therapies. This review provides a systematic summary of the role of ADMSCs in the treatment of central nervous system (CNS) disorders and explores their therapeutic potential for clinical application. ADMSC-related therapy offers a promising avenue to mitigate damage and enhance neurological function in central nervous system (CNS) disorders. However, further research is necessary to establish the safety and efficacy of clinical ADMSC-based therapy, optimize targeting accuracy, and refine delivery approaches for practical applications.
Collapse
Affiliation(s)
- Haiyue Wu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China; (H.W.); (Y.F.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yishu Fan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China; (H.W.); (Y.F.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Mengqi Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China; (H.W.); (Y.F.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
6
|
Choi J, Hwang J, Ramalingam M, Jeong HS, Jang S. Effects of HDAC inhibitors on neuroblastoma SH-SY5Y cell differentiation into mature neurons via the Wnt signaling pathway. BMC Neurosci 2023; 24:28. [PMID: 37127577 PMCID: PMC10152798 DOI: 10.1186/s12868-023-00798-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 04/19/2023] [Indexed: 05/03/2023] Open
Abstract
Histone deacetylase (HDAC) inhibitors affect cell homeostasis, gene expression, and cell cycle progression and promote cell terminal differentiation or apoptosis. However, the effect of HDAC inhibition on SH-SY5Y cells, which are neuroblastoma cells capable of differentiating into neurons under specific conditions, such as in the presence of retinoic acid (RA), is unknown. In this study, we hypothesized that HDAC inhibitors induced the neuronal differentiation of SH-SY5Y cells. To test this hypothesis, we used phase contrast microscopy, immunocytochemistry (ICC), qPCR, and western blotting analysis. MS-275 and valproic acid (VPA), two HDAC inhibitors, were selected to evaluate neuronal differentiation. It was confirmed that cells treated with MS-275 or VPA differentiated into mature neurons, which were distinguished by bipolar or multipolar morphologies with elongated branches. In addition, the mRNA expression of neuronal markers (Tuj1 and NEFH) and the oligodendrocyte marker (CNP) was significantly increased with MS-275 or VPA treatment compared to that with RA treatment. In addition, the protein expression of the other neuronal markers, Tuj1 and NeuN, was highly increased with HDAC inhibitor treatments compared to that with RA treatment. Furthermore, we confirmed that noncanonical Wnt signaling was upregulated by HDAC inhibitors via MAPK signaling and the Wnt/JNK pathway. Therefore, both MS-275 and VPA promoted the differentiation of SH-SY5Y cells into mature neurons via the Wnt signaling pathway.
Collapse
Affiliation(s)
- Jiyun Choi
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun, Jellanamdo, 58128, Republic of Korea
| | - Jinsu Hwang
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun, Jellanamdo, 58128, Republic of Korea
| | - Mahesh Ramalingam
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun, Jellanamdo, 58128, Republic of Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun, Jellanamdo, 58128, Republic of Korea.
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun, Jellanamdo, 58128, Republic of Korea.
| |
Collapse
|
7
|
Neuroprotective Effects of the Neural-Induced Adipose-Derived Stem Cell Secretome against Rotenone-Induced Mitochondrial and Endoplasmic Reticulum Dysfunction. Int J Mol Sci 2023; 24:ijms24065622. [PMID: 36982698 PMCID: PMC10054666 DOI: 10.3390/ijms24065622] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have therapeutic effects on neurodegenerative diseases (NDDs) known by their secreted molecules, referred to as the “secretome”. The mitochondrial complex I inhibitor, rotenone (ROT), reproduces α-synuclein (α-syn) aggregation seen in Parkinson’s disease (PD). In this present study, we examined the neuroprotective effects of the secretome from neural-induced human adipose tissue-derived stem cells (NI-ADSC-SM) during ROT toxicity in SH-SY5Y cells. Exposure to ROT significantly impaired the mitophagy by increased LRRK2, mitochondrial fission, and endoplasmic reticulum (ER) stress (ERS). ROT also increased the levels of calcium (Ca2+), VDAC, and GRP75, and decreased phosphorylated (p)-IP3R Ser1756/total (t)-IP3R1. However, NI-ADSC-SM treatment decreased Ca2+ levels along with LRRK2, insoluble ubiquitin, mitochondrial fission by halting p-DRP1 Ser616, ERS by reducing p-PERK Thr981, p-/t-IRE1α, p-SAPK, ATF4, and CHOP. In addition, NI-ADSC-SM restored the mitophagy, mitochondrial fusion, and tethering to the ER. These data suggest that NI-ADSC-SM decreases ROT-induced dysfunction in mitochondria and the ER, which subsequently stabilized tethering in mitochondria-associated membranes in SH-SY5Y cells.
Collapse
|
8
|
Ramalingam M, Jeong HS, Hwang J, Cho HH, Kim BC, Kim E, Jang S. Autophagy Signaling by Neural-Induced Human Adipose Tissue-Derived Stem Cell-Conditioned Medium during Rotenone-Induced Toxicity in SH-SY5Y Cells. Int J Mol Sci 2022; 23:ijms23084193. [PMID: 35457010 PMCID: PMC9031864 DOI: 10.3390/ijms23084193] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 12/04/2022] Open
Abstract
Rotenone (ROT) inhibits mitochondrial complex I, leading to reactive oxygen species formation, which causes neurodegeneration and alpha-synuclein (α-syn) aggregation and, consequently, Parkinson’s disease. We previously found that a neurogenic differentiated human adipose tissue-derived stem cell-conditioned medium (NI-hADSC-CM) was protective against ROT-induced toxicity in SH-SY5Y cells. In the present study, ROT significantly decreased the phospho (p)-mTORC1/total (t)-mTOR, p-mTORC2/t-mTOR, and p-/t-ULK1 ratios and the ATG13 level by increasing the DEPTOR level and p-/t-AMPK ratio. Moreover, ROT increased the p-/t-Akt ratio and glycogen synthase kinase-3β (GSK3β) activity by decreasing the p-/t-ERK1/2 ratios and beclin-1 level. ROT also promoted the lipidation of LC3B-I to LC3B-II by inducing autophagosome formation in Triton X-100-soluble and -insoluble cell lysate fractions. Additionally, the levels of ATG3, 5, 7, and 12 were decreased, along with those of lysosomal LAMP1, LAMP2, and TFEB, leading to lysosomal dysfunction. However, NI-hADSC-CM treatment increased the p-mTORC1, p-mTORC2, p-ULK1, p-Akt, p-ERK1/2, ATG13, and beclin-1 levels and decreased the p-AMPK level and GSK3β activity in response to ROT-induced toxicity. Additionally, NI-hADSC-CM restored the LC3B-I level, increased the p62 level, and normalized the ATG and lysosomal protein amounts to control levels. Autophagy array revealed that the secreted proteins in NI-hADSC-CM could be crucial in the neuroprotection. Taken together, our results showed that the neuroprotective effects of NI-hADSC-CM on the autophagy signaling pathways could alleviate the aggregation of α-syn in Parkinson’s disease and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Mahesh Ramalingam
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Korea; (H.-S.J.); (J.H.)
- Correspondence: (M.R.); (S.J.)
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Korea; (H.-S.J.); (J.H.)
| | - Jinsu Hwang
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Korea; (H.-S.J.); (J.H.)
| | - Hyong-Ho Cho
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Korea;
| | - Byeong C. Kim
- Department of Neurology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Korea;
| | - Eungpil Kim
- Jeonnam Biopharmaceutical Research Center, Hwasun 58141, Korea;
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Korea; (H.-S.J.); (J.H.)
- Correspondence: (M.R.); (S.J.)
| |
Collapse
|
9
|
Jang S, Hwang J, Jeong HS. The Role of Histone Acetylation in Mesenchymal Stem Cell Differentiation. Chonnam Med J 2022; 58:6-12. [PMID: 35169553 PMCID: PMC8813658 DOI: 10.4068/cmj.2022.58.1.6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 11/06/2022] Open
Abstract
The mechanism and action concerning epigenetic modifications, especially that of histone modifications, are not fully understood. However, it is clear that histone modifications play an essential role in several biological processes that are involved in cell proliferation and differentiation. In this article, we focused on how histone acetylation may result in differentiation into mesenchymal stem cells as well as histone acetylation function. Moreover, histone acetylation followed by the action of histone deacetylase inhibitors, which can result in the differentiation of stem cells into other types of cells such as adipocytes, chondrocytes, osteocytes, neurons, and other lineages, were also reviewed.
Collapse
Affiliation(s)
- Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun, Korea
| | - Jinsu Hwang
- Department of Physiology, Chonnam National University Medical School, Hwasun, Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
10
|
Gupta A, Singh S. Potential Role of Growth Factors Controlled Release in Achieving Enhanced Neuronal Trans-differentiation from Mesenchymal Stem Cells for Neural Tissue Repair and Regeneration. Mol Neurobiol 2021; 59:983-1001. [PMID: 34816381 DOI: 10.1007/s12035-021-02646-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
With an increase in the incidence of neurodegenerative diseases, a need to replace incapable conventional methods has arisen. To overcome this burden, stem cells therapy has emerged as an efficient treatment option. Endeavours to accomplish this have paved the path to neural regeneration through efficient neuronal transdifferentiation. Despite their potential, the use of stem cells still entails several limitations, such as low differentiation efficiency and difficulties in guiding differentiation. The process of neural differentiation through the stem cells is achieved through the use of chemical inducers or growth factors and their direct introduction reduces their bioavailability in the system. To address these limitations, neural regeneration ventures require growth factors to be effectively implemented on stem cells in order to produce functional neuronal precursor cells. An efficient technique to achieve it is through the delivery of growth factors via microcarriers for their sustained release. It ensures the presence of commensurable concentration even at later stages of neuronal transdifferentiation. Nanofibers and nanoparticles, along with liposomes and such, have been used to implement this. The interaction between such carriers and the growth factors is mainly electrostatic. Such interaction enables them to form a stable assembly through immobilisation of the growth factor either onto their surfaces or within the core of their structures. The rate of sustained release depends upon the release kinetics associated with the polymeric structure employed and its interaction with the encapsulated growth factor. The sustained release ensures that the stem cells immerse under the effect of the growth factors for a prolonged period, ultimately aiding in the formation of cells showing ample characteristics of neuron precursors. This review analyses the various carriers that have been employed for the release of growth factors in an orderly fashion and their constituents, along with the advantages and the limitations they pose in delivering the growth factors for facilitating the process of neuronal transdifferentiation.
Collapse
Affiliation(s)
- Ayushi Gupta
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India
| | - Sangeeta Singh
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India.
| |
Collapse
|
11
|
Zhao CG, Qin J, Li J, Jiang S, Ju F, Sun W, Ren Z, Ji YQ, Wang R, Sun XL, Mou X, Yuan H. LINGO-1 regulates Wnt5a signaling during neural stem and progenitor cell differentiation by modulating miR-15b-3p levels. Stem Cell Res Ther 2021; 12:372. [PMID: 34187584 PMCID: PMC8243903 DOI: 10.1186/s13287-021-02452-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/07/2021] [Indexed: 11/22/2022] Open
Abstract
Background Manipulation of neural stem and progenitor cells (NSPCs) is critical for the successful treatment of spinal cord injury (SCI) by NSPC transplantation, since their differentiation into neurons and oligodendrocytes can be inhibited by factors present in inflamed myelin. In this study, we examined the effects of LINGO-1 on spinal cord-derived NSPC (sp-NSPC) differentiation, the underlying mechanisms of action, and the functional recovery of mice after transplantation of manipulated cells. Methods sp-NSPCs were harvested from female adult C57/BL6 mice after SCI induced with an NYU impactor. These cells were infected with lentiviral vectors containing LINGO-1 shRNA sequence or a scrambled control and transplanted into SCI mice. Tuj-1- and GFAP-positive cells were assessed by immunofluorescence staining. Wnt5a, p-JNK, JNK, and β-catenin expression was determined by Western blot and RT-qPCR. miRNAs were sequenced to detect changes in miRNA expression. Motor function was evaluated 0–35 days post-surgery by means of the Basso Mouse Scale (BMS) and by the rotarod performance test. Results We discovered that LINGO-1 shRNA increased neuronal differentiation of sp-NSPCs while decreasing astrocyte differentiation. These effects were accompanied by elevated Wnt5a protein expression, but unexpectedly, no changes in Wnt5a mRNA levels. miRNA-sequence analysis demonstrated that miR-15b-3p was a downstream mediator of LINGO-1 which suppressed Wnt5a expression. Transplantation of LINGO-1 shRNA-treated sp-NSPCs into SCI mice promoted neural differentiation, wound compaction, and motor function recovery. Conclusions LINGO-1 shRNA promotes neural differentiation of sp-NSPCs and Wnt5a expression, probably by downregulating miR-15b-3p. Transplantation of LINGO-1 shRNA-treated NSPCs promotes recovery of motor function after SCI, highlighting its potential as a target for SCI treatment.
Collapse
Affiliation(s)
- Chen-Guang Zhao
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Qin
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jia Li
- Department of Medicine and Health, University Bretagne Occidentale, Brest, France
| | - Shan Jiang
- Department of Rehabilitation Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Fen Ju
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei Sun
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhen Ren
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu-Qiang Ji
- Department of Central Laboratory, The First Hospital of Xi'an, Xi'an, China
| | - Rui Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao-Long Sun
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiang Mou
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hua Yuan
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
12
|
Eftekhari BS, Eskandari M, Janmey PA, Samadikuchaksaraei A, Gholipourmalekabadi M. Conductive chitosan/polyaniline hydrogel with cell-imprinted topography as a potential substrate for neural priming of adipose derived stem cells. RSC Adv 2021; 11:15795-15807. [PMID: 35481217 PMCID: PMC9029165 DOI: 10.1039/d1ra00413a] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Biophysical characteristics of engineered scaffolds such as topography and electroconductivity have shown potentially beneficial effects on stem cell morphology, proliferation, and differentiation toward neural cells. In this study, we fabricated a conductive hydrogel made from chitosan (CS) and polyaniline (PANI) with induced PC12 cell surface topography using a cell imprinting technique to provide both topographical properties and conductivity in a platform. The engineered hydrogel's potential for neural priming of rat adipose-derived stem cells (rADSCs) was determined in vitro. The biomechanical analysis revealed that the electrical conductivity, stiffness, and hydrophobicity of flat (F) and cell-imprinted (CI) substrates increased with increased PANI content in the CS/PANI scaffold. The conductive substrates exhibited a lower degradation rate compared to non-conductive substrates. According to data obtained from F-actin staining and AFM micrographs, both CI(CS) and CI(CS-PANI) substrates induced the morphology of rADSCs from their irregular shape (on flat substrates) into the elongated and bipolar shape of the neuronal-like PC12 cells. Immunostaining analysis revealed that both CI(CS) and CI (CS-PANI) significantly upregulated the expression of GFAP and MAP2, two neural precursor-specific genes, in rADSCs compared with flat substrates. Although the results reveal that both cell-imprinted topography and electrical conductivity affect the neural lineage differentiation, some data demonstrate that the topography effects of the cell-imprinted surface have a more critical role than electrical conductivity on neural priming of ADSCs. The current study provides new insight into the engineering of scaffolds for nerve tissue engineering.
Collapse
Affiliation(s)
- Behnaz Sadat Eftekhari
- Department of Biomedical Engineering, Amirkabir University of Technology 424 Hafez Ave Tehran 15875-4413 Iran +98 21 6454 23 62
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania 1010 Vagelos Research Laboratories, 3340 Smith Walk Philadelphia PA 19104-6383 USA +1 215 573 6815 +1 215 573 7380
| | - Mahnaz Eskandari
- Department of Biomedical Engineering, Amirkabir University of Technology 424 Hafez Ave Tehran 15875-4413 Iran +98 21 6454 23 62
| | - Paul A Janmey
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania 1010 Vagelos Research Laboratories, 3340 Smith Walk Philadelphia PA 19104-6383 USA +1 215 573 6815 +1 215 573 7380
| | | | - Mazaher Gholipourmalekabadi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences Tehran Iran
- Cellular and Molecular Research Centre, Iran University of Medical Sciences Tehran Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences Tehran Iran
| |
Collapse
|
13
|
Rogan MR, Patterson LL, Byerly CD, Luo T, Paessler S, Veljkovic V, Quade B, McBride JW. Ehrlichia chaffeensis TRP120 Is a Wnt Ligand Mimetic That Interacts with Wnt Receptors and Contains a Novel Repetitive Short Linear Motif That Activates Wnt Signaling. mSphere 2021; 6:6/2/e00216-21. [PMID: 33883266 PMCID: PMC8546699 DOI: 10.1128/msphere.00216-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ehrlichia chaffeensis expresses the TRP120 multifunctional effector, which is known to play a role in phagocytic entry, on the surface of infectious dense-cored ehrlichiae, but a cognate host receptor has not been identified. We recently reported that E. chaffeensis activates canonical Wnt signaling in monocytes to promote bacterial uptake and intracellular survival and that TRP120 was involved in this activation event. To identify the specific mechanism of pathway activation, we hypothesized that TRP120 is a Wnt signaling ligand mimetic that initiates Wnt pathway activity through direct interaction with the Wnt pathway Frizzled family of receptors. In this study, we used confocal immunofluorescence microscopy to demonstrate very strong colocalization between E. chaffeensis and Fzd2, 4, 5, 7, and 9 as well as coreceptor LRP5 at 1 to 3 h postinfection. Direct binding between TRP120 and multiple Fzd receptors was further confirmed by enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (SPR). Interfering RNA knockdown of Wnt receptors, coreceptors, and signaling pathway components significantly reduced E. chaffeensis infection, demonstrating that complex and redundant interactions are involved in Wnt pathway exploitation. We utilized in silico approaches to identify a repetitive short linear motif (SLiM) in TRP120 that is homologous to Wnt ligands and used mutant SLiM peptides and an α-TRP120-Wnt-SLiM antibody to demonstrate that the TRP120 Wnt SLiM activates the canonical Wnt pathway and promotes E. chaffeensis infection. This study reports the first example of bacterial mimicry of Wnt pathway ligands and highlights a pathogenic mechanism with potential for targeting by antimicrobial therapeutics.IMPORTANCE Upon infecting mammalian hosts, Ehrlichia chaffeensis establishes a replicative niche in microbe-eating immune system cells where it expertly orchestrates infection and spread. One of the ways Ehrlichia survives within these phagocytes is by activating evolutionarily conserved signaling pathways including the Wnt pathway; however, the molecular details of pathway hijacking have not been defined. This study is significant because it identifies an ehrlichial protein that directly interacts with components of the Wnt receptor complex, influencing pathway activity and promoting infection. Consequentially, Ehrlichia serves as a unique tool to investigate the intricacies of how pathogens repurpose human immune cell signaling and provides an opportunity to better understand many cellular processes in health and disease. Furthermore, understanding how this bacterium utilizes its small genome to survive within cells that evolved to destroy pathogens will facilitate the development of antibacterial therapeutics that could target Ehrlichia as well as other intracellular agents of human disease.
Collapse
Affiliation(s)
- Madison R Rogan
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - LaNisha L Patterson
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Caitlan D Byerly
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Tian Luo
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- BiomedProtection, LLC, Galveston, Texas, USA
| | | | - Bethany Quade
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jere W McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
14
|
de los Reyes Corrales T, Losada-Pérez M, Casas-Tintó S. JNK Pathway in CNS Pathologies. Int J Mol Sci 2021; 22:3883. [PMID: 33918666 PMCID: PMC8070500 DOI: 10.3390/ijms22083883] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 12/13/2022] Open
Abstract
The c-Jun N-terminal kinase (JNK) signalling pathway is a conserved response to a wide range of internal and external cellular stress signals. Beside the stress response, the JNK pathway is involved in a series of vital regulatory mechanisms during development and adulthood that are critical to maintain tissue homeostasis. These mechanisms include the regulation of apoptosis, growth, proliferation, differentiation, migration and invasion. The JNK pathway has a diverse functionality and cell-tissue specificity, and has emerged as a key player in regeneration, tumorigenesis and other pathologies. The JNK pathway is highly active in the central nervous system (CNS), and plays a central role when cells need to cope with pathophysiological insults during development and adulthood. Here, we review the implications of the JNK pathway in pathologies of the CNS. More specifically, we discuss some newly identified examples and mechanisms of JNK-driven tumor progression in glioblastoma, regeneration/repair after an injury, neurodegeneration and neuronal cell death. All these new discoveries support the central role of JNK in CNS pathologies and reinforce the idea of JNK as potential target to reduce their detrimental effects.
Collapse
|
15
|
Ramalingam M, Jang S, Jeong HS. Neural-Induced Human Adipose Tissue-Derived Stem Cells Conditioned Medium Ameliorates Rotenone-Induced Toxicity in SH-SY5Y Cells. Int J Mol Sci 2021; 22:ijms22052322. [PMID: 33652595 PMCID: PMC7956615 DOI: 10.3390/ijms22052322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is an age-related neurodegenerative disease (NDD) characterized by the degenerative loss of dopaminergic neurons in the substantia nigra along with aggregation of α-synuclein (α-syn). Neurogenic differentiation of human adipose-derived stem cells (NI-hADSCs) by supplementary factors for 14 days activates different biological signaling pathways. In this study, we evaluated the therapeutic role of NI-hADSC-conditioned medium (NI-hADSC-CM) in rotenone (ROT)-induced toxicity in SH-SY5Y cells. Increasing concentrations of ROT led to decreased cell survival at 24 and 48 h in a dose- and time-dependent manner. Treatment of NI-hADSC-CM (50% dilution in DMEM) against ROT (0.5 μM) significantly increased the cell survival. ROT toxicity decreased the expression of tyrosine hydroxylase (TH). Western blot analysis of the Triton X-100-soluble fraction revealed that ROT significantly decreased the oligomeric, dimeric, and monomeric phosphorylated Serine129 (p-S129) α-syn, as well as the total monomeric α-syn expression levels. ROT toxicity increased the oligomeric, but decreased the dimeric and monomeric p-S129 α-syn expression levels. Total α-syn expression (in all forms) was increased in the Triton X-100-insoluble fraction, compared to the control. NI-hADSC-CM treatment enhanced the TH expression, stabilized α-syn monomers, reduced the levels of toxic insoluble p-S129 α-syn, improved the expression of neuronal functional proteins, regulated the Bax/Bcl-2 ratio, and upregulated the expression of pro-caspases, along with PARP-1 inactivation. Moreover, hADSC-CM treatment decreased the cell numbers and have no effect against ROT toxicity on SH-SY5Y cells. The therapeutic effects of NI-hADSC-CM was higher than the beneficial effects of hADSC-CM on cellular signaling. From these results, we conclude that NI-hADSC-CM exerts neuroregenerative effects on ROT-induced PD-like impairments in SH-SY5Y cells.
Collapse
|
16
|
Therapeutic Effects of Conditioned Medium of Neural Differentiated Human Bone Marrow-Derived Stem Cells on Rotenone-Induced Alpha-Synuclein Aggregation and Apoptosis. Stem Cells Int 2021; 2021:6658271. [PMID: 33552161 PMCID: PMC7847328 DOI: 10.1155/2021/6658271] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/29/2020] [Accepted: 01/05/2021] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been used against several diseases. Their potential mainly appears from its secreted biomolecules. Human bone marrow-derived stem cells (hBMSC) displayed neuronal functional characteristics after differentiation by basic fibroblast growth factor (bFGF) and forskolin. PD is a chronic age-related neurodegenerative disease (NDD) characterized by loss of dopaminergic neurons in the substantia nigra (SN) and abnormal accumulation of α-synuclein (α-syn) aggregations. In this present study, we evaluated the therapeutic effects of neural differentiated hBMSC (NI-hBMSC) conditioned medium (NI-hBMSC-CM) to a rotenone- (ROT-) induced Parkinson's disease (PD) model in SH-SY5Y cells. NI-hBMSC-CM treatment (50% diluted) in the last 24 h of 48 h ROT (0.5 μM) toxicity showed a significant increase in cell survival. The decreased tyrosine hydroxylase (TH) expression as a hallmark of PD was increased by NI-hBMSC-CM. The Triton X-100-soluble and Triton X-100-insoluble cell lysate fractions were used in Western blotting. The oligomeric, dimeric, and monomeric phosphorylated serine129 (p-S129) α-syn and total monomeric α-syn were decreased during ROT toxicity in the Triton X-100-soluble fraction. The Triton X-100-insoluble fraction revealed that ROT toxicity significantly increased the oligomeric but decreased the dimeric and monomeric p-S129 α-syn expressions while all forms of total α-syn were increased in SH-SY5Y cells. NI-hBMSC-CM stabilized the physiological α-syn monomers and reduced aggregated insoluble p-S129 α-syn against ROT. The cytoskeletal proteins, neurofilament-H (NF-H), β3-tubulin (Tuj1), neuronal nuclei (NeuN), and synaptophysin (SYP) were significantly decreased during ROT toxicity. In addition, proapoptotic Bax was increased by ROT with decreased antiapoptotic Bcl-2 and Mcl-1 as well as proforms of caspase-9, caspase-3, caspase-7, and PARP-1. NI-hBMSC-CM ameliorated the neurotrophic protein expressions, controlled the Bax/Bcl-2 ratio, upregulated procaspases, and inactivated PARP-1. From our results, we conclude that NI-hBMSC-CM containing released biomolecules during neural differentiation employs regenerative effects on the ROT model of PD in SH-SY5Y cells.
Collapse
|
17
|
Transplantation of Wnt5a-modified NSCs promotes tissue repair and locomotor functional recovery after spinal cord injury. Exp Mol Med 2020; 52:2020-2033. [PMID: 33311637 PMCID: PMC8080632 DOI: 10.1038/s12276-020-00536-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 09/15/2020] [Accepted: 10/14/2020] [Indexed: 01/15/2023] Open
Abstract
Traditional therapeutic strategies for spinal cord injury (SCI) are insufficient to repair locomotor function because of the failure of axonal reconnection and neuronal regeneration in the injured central nervous system (CNS). Neural stem cell (NSC) transplantation has been considered a potential strategy and is generally feasible for repairing the neural circuit after SCI; however, the most formidable problem is that the neuronal differentiation rate of NSCs is quite limited. Therefore, it is essential to induce the neuronal differentiation of NSCs and improve the differentiation rate of NSCs in spinal cord repair. Our results demonstrate that both Wnt5a and miRNA200b-3p could promote NSC differentiation into neurons and that Wnt5a upregulated miRNA200b-3p expression through MAPK/JNK signaling to promote NSC differentiation into neurons. Wnt5a could reduce RhoA expression by upregulating miRNA200b-3p expression to inhibit activation of the RhoA/Rock signaling pathway, which has been reported to suppress neuronal differentiation. Overexpression of RhoA abolished the neurogenic capacity of Wnt5a and miRNA200b-3p. In vivo, miRNA200b-3p was critical for Wnt5a-induced NSC differentiation into neurons to promote motor functional and histological recovery after SCI by suppressing RhoA/Rock signaling. These findings provide more insight into SCI and help with the identification of novel treatment strategies. Incorporating key molecules into neural stem cells enhances their ability to differentiate correctly and promote repair following spinal cord injury. Spinal cord injuries can have a debilitating effect on patients’ lives, yet there are no therapies that fully restore movement and sensation. Therapies based on neural stem cells (NSCs) show promise, but initial studies show many NSCs differentiate into astrocytes, supportive cells that do not conduct nerve impulses, instead of neurons, leading to treatment failure. Yong Wan and Le Wang at Sun Yat-sen University in Guangzhou, China, and co-workers demonstrated that adding a protein called Wnt5a and a specific microRNA molecule to NSCs significantly increases differentiation into neurons. Wnt5a suppresses a signalling pathway that otherwise interferes with NSC differentiation. Experiments on rat models showed that the therapy improved locomotor function and tissue repair after injury.
Collapse
|
18
|
Mitra S, Kaushik N, Moon IS, Choi EH, Kaushik NK. Utility of Reactive Species Generation in Plasma Medicine for Neuronal Development. Biomedicines 2020; 8:E348. [PMID: 32932745 PMCID: PMC7555638 DOI: 10.3390/biomedicines8090348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are critical signaling molecules for neuronal physiology that stimulate growth and development and play vital roles in several pathways when in a balanced state, but they cause neurodegeneration when unbalanced. As ROS levels above a certain threshold cause the activation of the autophagy system, moderate levels of ROS can be used as treatment strategies. Currently, such treatments are used together with low-level laser or photodynamic therapies, photo-bio modulation, or infrared treatments, in different chronic diseases but not in the treatment of neurodegeneration. Recently, non-thermal plasma has been successfully used in biomedical applications and treatments, and beneficial effects such as differentiation, cell growth, and proliferation, stimulation of ROS based pathways have been observed. Besides the activation of a wide range of biological signaling pathways by generating ROS, plasma application can be an effective treatment in neuronal regeneration, as well as in neuronal diseases. In this review, we summarize the generation and role of ROS in neurons and provide critical insights into their potential benefits on neurons. We also discuss the underlying mechanisms of ROS on neuronal development. Regarding clinical applications, we focus on ROS-based neuronal growth and regeneration strategies and in the usage of non-thermal plasma in neuronal and CNS injury treatments.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Plasma Bioscience Research Center/Applied Plasma Medicine Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea;
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea;
| | - Neha Kaushik
- Department of Biotechnology, University of Suwon, Hwaseong 18323, Korea;
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea;
| | - Eun Ha Choi
- Plasma Bioscience Research Center/Applied Plasma Medicine Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea;
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center/Applied Plasma Medicine Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea;
| |
Collapse
|
19
|
McClintick JN, Thapa K, Liu Y, Xuei X, Edenberg HJ. Effects of chronic intermittent ethanol exposure and withdrawal on neuroblastoma cell transcriptome. Alcohol 2020; 85:119-126. [PMID: 31923563 PMCID: PMC7237278 DOI: 10.1016/j.alcohol.2019.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/08/2019] [Accepted: 12/22/2019] [Indexed: 02/06/2023]
Abstract
Cycles of heavy drinking and abstinence can lead to alcohol use disorder. We studied the effects of chronic intermittent ethanol exposure (CIE) over 3 weeks on neuroblastoma cells, using an ethanol concentration frequently attained in binge drinking (40 mM, 184 mg/dL). There were many changes in gene expression but most were small. CIE affected pathways instrumental in the development or plasticity of neurons, including axonal guidance, reelin signaling, and synaptogenesis. Genes involved in dopamine and serotonin signaling were also affected. Changes in transporters and receptors could dampen both NMDA and norepinephrine transmissions. Decreased expression of the GABA transporter SLC6A11 could increase GABA transmission and has been associated with a switch from sweet drinking to ethanol consumption in rats. Ethanol increased stress responses such as the unfolded protein response. TGF-β and NFκB signaling were increased. Most of the genes involved in cholesterol biosynthesis were decreased in expression. Withdrawal for 24 h after CIE caused most of the CIE-induced expression changes to move back toward unexposed levels.
Collapse
Affiliation(s)
- Jeanette N McClintick
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kriti Thapa
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiaoling Xuei
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
20
|
Gao J, Liao Y, Qiu M, Shen W. Wnt/β-Catenin Signaling in Neural Stem Cell Homeostasis and Neurological Diseases. Neuroscientist 2020; 27:58-72. [PMID: 32242761 DOI: 10.1177/1073858420914509] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neural stem/progenitor cells (NSCs) maintain the ability of self-renewal and differentiation and compose the complex nervous system. Wnt signaling is thought to control the balance of NSC proliferation and differentiation via the transcriptional coactivator β-catenin during brain development and adult tissue homeostasis. Disruption of Wnt signaling may result in developmental defects and neurological diseases. Here, we summarize recent findings of the roles of Wnt/β-catenin signaling components in NSC homeostasis for the regulation of functional brain circuits. We also suggest that the potential role of Wnt/β-catenin signaling might lead to new therapeutic strategies for neurological diseases, including, but not limited to, spinal cord injury, Alzheimer's disease, Parkinson's disease, and depression.
Collapse
Affiliation(s)
- Juanmei Gao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China.,College of Life and Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuan Liao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China.,College of Life and Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wanhua Shen
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
21
|
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
|
22
|
Li X, Peng Z, Long L, Tuo Y, Wang L, Zhao X, Le W, Wan Y. Wnt4-modified NSC transplantation promotes functional recovery after spinal cord injury. FASEB J 2020; 34:82-94. [PMID: 31914702 DOI: 10.1096/fj.201901478rr] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/27/2019] [Accepted: 10/08/2019] [Indexed: 01/26/2023]
Abstract
Spinal cord injury (SCI) can lead to severe motor and sensory dysfunction, yet there are no effective therapies currently due to the failure of reconstructing the interruption of the neuroanatomical circuit. While neural stem cell (NSC) transplantation has been considered a potential strategy to repair the neural circuit after SCI, the efficacy of this strategy remains unproven. The main reason is that most of the transplanted NSC differentiates into astrocyte rather than neuron in the microenvironment of SCI. Our results demonstrated that Wnt4 significantly promotes the differentiation of NSC into neuron by activating both β-catenin and MAPK/JNK pathways and suppressing the activation of Notch signaling, which is acknowledged as prevention of NSC differentiation into neuron, through downregulating NICD expression, translocating and preventing the combination of NICD and RbpJ in nucleus. In addition, Wnt4 rescues the negative effect of Jagged, the ligand of Notch signaling, to promote neuronal differentiation. Moreover, in vivo study, transplantation of Wnt4-modified NSC efficaciously repairs the injured spinal cord and recovers the motor function of hind limbs after SCI. This study sheds new light into mechanisms that Wnt4-modified NSC transplantation is sufficient to repair the injured spinal cord and recover the motor dysfunction after SCI.
Collapse
Affiliation(s)
- Xiang Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiming Peng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lingli Long
- Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Tuo
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liqin Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyang Zhao
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wang Le
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- The First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Yong Wan
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- The First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| |
Collapse
|
23
|
George S, Hamblin MR, Abrahamse H. Differentiation of Mesenchymal Stem Cells to Neuroglia: in the Context of Cell Signalling. Stem Cell Rev Rep 2019; 15:814-826. [PMID: 31515658 PMCID: PMC6925073 DOI: 10.1007/s12015-019-09917-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The promise of engineering specific cell types from stem cells and rebuilding damaged or diseased tissues has fascinated stem cell researchers and clinicians over last few decades. Mesenchymal Stem Cells (MSCs) have the potential to differentiate into non-mesodermal cells, particularly neural-lineage, consisting of neurons and glia. These multipotent adult stem cells can be used for implementing clinical trials in neural repair. Ongoing research identifies several molecular mechanisms involved in the speciation of neuroglia, which are tightly regulated and interconnected by various components of cell signalling machinery. Growing MSCs with multiple inducers in culture media will initiate changes on intricately interlinked cell signalling pathways and processes. Net result of these signal flow on cellular architecture is also dependent on the type of ligands and stem cells investigated in vitro. However, our understanding about this dynamic signalling machinery is limited and confounding, especially with spheroid structures, neurospheres and organoids. Therefore, the results for differentiating neurons and glia in vitro have been inconclusive, so far. Added to this complication, we have no convincing evidence about the electrical conductivity and functionality status generated in differentiating neurons and glia. This review has taken a step forward to tailor the information on differentiating neuroglia with the common methodologies, in practice.
Collapse
Affiliation(s)
- Sajan George
- Laser Research Centre, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028, South Africa
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028, South Africa
- Wellman Centre for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Heidi Abrahamse
- Laser Research Centre, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028, South Africa.
| |
Collapse
|
24
|
Adipose Stem Cell-Based Clinical Strategy for Neural Regeneration: A Review of Current Opinion. Stem Cells Int 2019; 2019:8502370. [PMID: 31827536 PMCID: PMC6885831 DOI: 10.1155/2019/8502370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/02/2019] [Accepted: 11/11/2019] [Indexed: 02/05/2023] Open
Abstract
Nerve injury is a critical problem in the clinic. Nerve injury causes serious clinic issues including pain and dysfunctions for patients. The disconnection between damaged neural fibers and muscles will result in muscle atrophy in a few weeks if no treatment is applied. Moreover, scientists have discovered that nerve injury can affect the osteogenic differentiation of skeletal stem cells (SSCs) and the fracture repairing. In plastic surgery, muscle atrophy and bone fracture after nerve injury have plagued clinicians for many years. How to promote neural regeneration is the core issue of research in the recent years. Without obvious effects of traditional neurosurgical treatments, research on stem cells in the past 10 years has provided a new therapeutic strategy for us to address this problem. Adipose stem cells (ASCs) are a kind of mesenchymal stem cells that have differentiation potential in adipose tissue. In the recent years, ASCs have become the focus of regenerative medicine. They play a pivotal role in tissue regeneration engineering. As a type of stem cell, ASCs are becoming popular for neuroregenerative medicine due to their advantages and characteristics. In the various diseases of the nervous system, ASCs are gradually applied to treat the related diseases. This review article focuses on the mechanism and clinical application of ASCs in nerve regeneration as well as the related research on ASCs over the past decades.
Collapse
|
25
|
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
|
26
|
Chu DT, Nguyen Thi Phuong T, Tien NLB, Tran DK, Minh LB, Thanh VV, Gia Anh P, Pham VH, Thi Nga V. Adipose Tissue Stem Cells for Therapy: An Update on the Progress of Isolation, Culture, Storage, and Clinical Application. J Clin Med 2019; 8:E917. [PMID: 31247996 PMCID: PMC6678927 DOI: 10.3390/jcm8070917] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/10/2019] [Accepted: 06/21/2019] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue stem cells (ASCs), known as multipotent stem cells, are most commonly used in the clinical applications in recent years. Adipose tissues (AT) have the advantage in the harvesting, isolation, and expansion of ASCs, especially an abundant amount of stem cells compared to bone marrow. ASCs can be found in stromal vascular fractions (SVF) which are easily obtained from the dissociation of adipose tissue. Both SVFs and culture-expanded ASCs exhibit the stem cell characteristics such as differentiation into multiple cell types, regeneration, and immune regulators. Therefore, SVFs and ASCs have been researched to evaluate the safety and benefits for human use. In fact, the number of clinical trials on ASCs is going to increase by years; however, most trials are in phase I and II, and lack phase III and IV. This systemic review highlights and updates the process of the harvesting, characteristics, isolation, culture, storage, and application of ASCs, as well as provides further directions on the therapeutic use of ASCs.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam.
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam.
| | - Thuy Nguyen Thi Phuong
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju 61186, Korea
| | - Nguyen Le Bao Tien
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam
| | - Dang Khoa Tran
- Department of Anatomy, University of Medicine Pham Ngoc Thach, Ho Chi Minh City 700000, Vietnam
| | - Le Bui Minh
- NTT Hi-tech Institute, Nguyen Tat Thanh University, 300A Nguyen Tat Thanh St., Ward 13, District 4, Ho Chi Minh City 700000, Vietnam
| | - Vo Van Thanh
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam
- Department of Surgery, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Pham Gia Anh
- Oncology Department, Viet Duc Hospital, Hanoi 100000, Vietnam
| | - Van Huy Pham
- AI Lab, Faculty of Information Technology, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
| | - Vu Thi Nga
- Institute for Research and Development, Duy Tan University, Danang 550000, Vietnam.
| |
Collapse
|
27
|
Zhang J, He L, Yang Z, Li L, Cai W. Lithium chloride promotes proliferation of neural stem cells in vitro, possibly by triggering the Wnt signaling pathway. Anim Cells Syst (Seoul) 2018; 23:32-41. [PMID: 30834157 PMCID: PMC6394309 DOI: 10.1080/19768354.2018.1487334] [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: 02/12/2018] [Accepted: 05/04/2018] [Indexed: 01/09/2023] Open
Abstract
The objective of this study was to clarify the relationship between the effect and associated mechanisms of lithium chloride on neural stem cells (NSCs) and the Wnt signaling pathway. The expression of key molecules proteins related to the Wnt signaling pathway in the proliferation and differentiation of control NSCs and lithium chloride-treated NSCs was detected by Western blot analysis. Flow cytometry analysis was applied to study the cell cycle dynamics of control NSCs and NSCs treated with lithium chloride. The therapeutic concentrations of lithium chloride stimulated NSC proliferation. β-catenin expression gradually decreased, while Gsk-3β expression gradually increased (P < 0.01). Furthermore, NSCs treated with lithium chloride showed significantly enhanced β-catenin expression and inhibited Gsk-3β expression in a dose-dependent manner. NSCs in the G0/G1-phases were activated with an increased therapeutic concentration of lithium chloride, while NSCs in the S-phase, as well as G2/M-phases, were arrested (P < 0.01). These data confirm that the proliferation of NSCs is remarkably promoted through changes of cell dynamics after treatment with lithium chloride. Our results provide insight into the effects of lithium chloride in promoting the proliferation abilities of NSCs in vitro and preventing the cells from differentiating, which is potentially mediated by activation of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Geriatrics, Chinese PLA 113rd Hospital, Ningbo, People's Republic of China
| | - Lu He
- Department of Geriatrics, Chinese PLA 113rd Hospital, Ningbo, People's Republic of China
| | - Zhong Yang
- Department of Neurobiology, The Third Military Medical University, Chongqing, People's Republic of China
| | - Lihong Li
- Department of Neurobiology, The Third Military Medical University, Chongqing, People's Republic of China
| | - Wenqin Cai
- Department of Neurobiology, The Third Military Medical University, Chongqing, People's Republic of China
| |
Collapse
|
28
|
Luo L, Hu DH, Yin JQ, Xu RX. Molecular Mechanisms of Transdifferentiation of Adipose-Derived Stem Cells into Neural Cells: Current Status and Perspectives. Stem Cells Int 2018; 2018:5630802. [PMID: 30302094 PMCID: PMC6158979 DOI: 10.1155/2018/5630802] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/12/2018] [Accepted: 07/19/2018] [Indexed: 12/19/2022] Open
Abstract
Neurological diseases can severely compromise both physical and psychological health. Recently, adult mesenchymal stem cell- (MSC-) based cell transplantation has become a potential therapeutic strategy. However, most studies related to the transdifferentiation of MSCs into neural cells have had disappointing outcomes. Better understanding of the mechanisms underlying MSC transdifferentiation is necessary to make adult stem cells more applicable to treating neurological diseases. Several studies have focused on adipose-derived stromal/stem cell (ADSC) transdifferentiation. The purpose of this review is to outline the molecular characterization of ADSCs, to describe the methods for inducing ADSC transdifferentiation, and to examine factors influencing transdifferentiation, including transcription factors, epigenetics, and signaling pathways. Exploring and understanding the mechanisms are a precondition for developing and applying novel cell therapies.
Collapse
Affiliation(s)
- Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi 710032, China
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China
- Bayi Brain Hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Da-Hai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi 710032, China
| | - James Q. Yin
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China
- Bayi Brain Hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Ru-Xiang Xu
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China
- Bayi Brain Hospital, General Hospital of PLA Army, Beijing 100700, China
| |
Collapse
|
29
|
Han C, Li J, Wang C, Ouyang H, Ding X, Liu Y, Chen S, Luo L. Wnt5a Contributes to the Differentiation of Human Embryonic Stem Cells into Lentoid Bodies Through the Noncanonical Wnt/JNK Signaling Pathway. ACTA ACUST UNITED AC 2018; 59:3449-3460. [DOI: 10.1167/iovs.18-23902] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Chenlu Han
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jinyan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Chunxiao Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Hong Ouyang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoyan Ding
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Shuyi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Lixia Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
30
|
Jang S, Jeong HS. Data for the effect of histone deacetylase inhibitors on voltage- and ligand-gated ion channel gene expression in neurogenic induced-human adipose tissue-derived mesenchymal stem cells. Data Brief 2018; 17:1314-1319. [PMID: 29876485 PMCID: PMC5988444 DOI: 10.1016/j.dib.2018.02.058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/22/2018] [Accepted: 02/22/2018] [Indexed: 12/18/2022] Open
Abstract
This data article contains descriptive and experimental data on ion channel gene expressions following the histone deacetylase (HDAC) inhibitor treatment of neural induced human adipose tissue-derived mesenchymal stem cells (NI-hADSCs). Following treatment of the HDAC inhibitors, such as MS-275, NaB, TSA, or VPA, the phenotypes of NI-hADSCs exhibit neuron-like features and the neurofilament-L (NFL)-positive cells were increased. The expression of the ion channel marker genes, such as SCN5A, KCNA4, and CACNA1G, was highly increased following treatment with the HDAC inhibitors; however, the expression of others was either decreased or unchanged. For further details and experimental findings please refer to the research article by Jang and Jeong. Histone deacetylase inhibition-mediated neuronal differentiation via the Wnt signaling pathway in human adipose tissue-derived mesenchymal stem cells (Jang and Jeong, 2018) [1].
Collapse
Affiliation(s)
- Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
31
|
Histone deacetylase inhibition-mediated neuronal differentiation via the Wnt signaling pathway in human adipose tissue-derived mesenchymal stem cells. Neurosci Lett 2018; 668:24-30. [DOI: 10.1016/j.neulet.2018.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/22/2017] [Accepted: 01/03/2018] [Indexed: 01/06/2023]
|
32
|
Jang S, Park SH. Antidiabetic Drug Metformin Protects Neuronal Cells against Quinolinic Acid-Induced Excitotoxicity by Decreasing Intracellular Calcium. Chonnam Med J 2018; 54:24-30. [PMID: 29399562 PMCID: PMC5794475 DOI: 10.4068/cmj.2018.54.1.24] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/19/2018] [Accepted: 01/20/2018] [Indexed: 12/19/2022] Open
Abstract
The antidiabetic drug metformin has been found to have beneficial effects in various neurological disorders; however, the molecular mechanisms underlying these effects remain unclear. Here we report that metformin protects neuronal cells from quinolinic acid (QUIN)-induced excitotoxicity. For this, we pretreated N18D3 neuronal cells with metformin prior to QUIN for 24 h. We found that pretreating the cells with metformin significantly improved cell survival rate in a concentration-dependent manner and reduced apoptotic cell death, as revealed by a MTT assay and DAPI staining, respectively. Calcium imaging using fluo-4 showed that metformin (100 µM) inhibited the intracellular calcium increase that was induced by QUIN. In addition, mRNA expression of pro-apoptotic genes, p21 and Bax, was decreased and of anti-apoptotic genes, Bcl-2 and Bcl-xl, was increased with metformin treatment compared to QUIN-induced cells. The immunoreactivity of phosphorylated ERK1/2 was elevated in cells treated with metformin, indicating the ERK1/2 signaling pathway in the neuroprotective effects of metformin in QUIN-induced cell death. Collectively, our data demonstrates that metformin exerts its neuroprotective effects by inhibiting intracellular calcium increases, allowing it to regulate ERK1/2 signaling and modulate cell survival and death genes.
Collapse
Affiliation(s)
- Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Gwangju, Korea.,Research Institute of Medical Sciences, Chonnam National University, Gwangju, Korea
| | - Sah-Hoon Park
- Department of Physiology, Chonnam National University Medical School, Gwangju, Korea.,Research Institute of Medical Sciences, Chonnam National University, Gwangju, Korea
| |
Collapse
|
33
|
Foulquier S, Daskalopoulos EP, Lluri G, Hermans KCM, Deb A, Blankesteijn WM. WNT Signaling in Cardiac and Vascular Disease. Pharmacol Rev 2018; 70:68-141. [PMID: 29247129 PMCID: PMC6040091 DOI: 10.1124/pr.117.013896] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
WNT signaling is an elaborate and complex collection of signal transduction pathways mediated by multiple signaling molecules. WNT signaling is critically important for developmental processes, including cell proliferation, differentiation and tissue patterning. Little WNT signaling activity is present in the cardiovascular system of healthy adults, but reactivation of the pathway is observed in many pathologies of heart and blood vessels. The high prevalence of these pathologies and their significant contribution to human disease burden has raised interest in WNT signaling as a potential target for therapeutic intervention. In this review, we first will focus on the constituents of the pathway and their regulation and the different signaling routes. Subsequently, the role of WNT signaling in cardiovascular development is addressed, followed by a detailed discussion of its involvement in vascular and cardiac disease. After highlighting the crosstalk between WNT, transforming growth factor-β and angiotensin II signaling, and the emerging role of WNT signaling in the regulation of stem cells, we provide an overview of drugs targeting the pathway at different levels. From the combined studies we conclude that, despite the sometimes conflicting experimental data, a general picture is emerging that excessive stimulation of WNT signaling adversely affects cardiovascular pathology. The rapidly increasing collection of drugs interfering at different levels of WNT signaling will allow the evaluation of therapeutic interventions in the pathway in relevant animal models of cardiovascular diseases and eventually in patients in the near future, translating the outcomes of the many preclinical studies into a clinically relevant context.
Collapse
Affiliation(s)
- Sébastien Foulquier
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Evangelos P Daskalopoulos
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Gentian Lluri
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Kevin C M Hermans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Arjun Deb
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| |
Collapse
|
34
|
Andrographolide Promotes Neural Differentiation of Rat Adipose Tissue-Derived Stromal Cells through Wnt/ β-Catenin Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4210867. [PMID: 29085837 PMCID: PMC5632471 DOI: 10.1155/2017/4210867] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/05/2017] [Accepted: 08/06/2017] [Indexed: 12/17/2022]
Abstract
Adipose tissue-derived stromal cells (ADSCs) are a high-yield source of pluripotent stem cells for use in cell-based therapies. We explored the effect of andrographolide (ANDRO, one of the ingredients of the medicinal herb extract) on the neural differentiation of rat ADSCs and associated molecular mechanisms. We observed that rat ADSCs were small and spindle-shaped and expressed multiple stem cell markers including nestin. They were multipotent as evidenced by adipogenic, osteogenic, chondrogenic, and neural differentiation under appropriate conditions. The proportion of cells exhibiting neural-like morphology was higher, and neurites developed faster in the ANDRO group than in the control group in the same neural differentiation medium. Expression levels of the neural lineage markers MAP2, tau, GFAP, and β-tubulin III were higher in the ANDRO group. ANDRO induced a concentration-dependent increase in Wnt/β-catenin signaling as evidenced by the enhanced expression of nuclear β-catenin and the inhibited form of GSK-3β (pSer9). Thus, this study shows for the first time how by enhancing the neural differentiation of ADSCs we expect that ANDRO pretreatment may increase the efficacy of adult stem cell transplantation in nervous system diseases, but more exploration is needed.
Collapse
|
35
|
Jang S, Cho HH, Park JS, Jeong HS. Non-canonical Wnt mediated neurogenic differentiation of human bone marrow-derived mesenchymal stem cells. Neurosci Lett 2017; 660:68-73. [PMID: 28916299 DOI: 10.1016/j.neulet.2017.09.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/24/2017] [Accepted: 09/11/2017] [Indexed: 12/23/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs), which are characterized by multipotency and self-renewal, are responsible for tissue regeneration and repair. We have previously reported in adipose tissue-derived MSCs that only Wnt5a is enhanced at neurogenic differentiation, and the mechanism of differentiation is dependent on the Wnt5a/JNK pathway; however, the role of Wnt/MAPK pathway is yet to be investigated in neurogenic differentiation in BM-MSCs. We compared the transcriptional expression of Wnt in neurogenic induced-hBM-MSCs (NI-hBM-MSCs) with that in primary hBM-MSCs, using RT-PCR, qPCR, and western blotting. Although the expression of Wnt1 and Wnt2 was unchanged, the expression of Wnt4, Wnt5a, and Wnt11 increased after neurogenic differentiation. In addition, only the expression of frizzled class receptor (Fzd) 3 gene was increased, but not of most of the Fzds and Wnt ligands in NI-hBM-MSCs. Interestingly, Wnt4, Wnt5a, and Wnt11 gene expressions significantly increased in NI-hBM-MSCs by qPCR. In addition, the protein expression level of Wnt4 and Wnt5a, but not Wnt3, increased after neurogenic induction. Furthermore, the expressions of phosphorylated-GSK-3β, ERK1/2, and PKC decreased; however, JNK was activated after neurogenic differentiation. Thus, non-canonical Wnts, i.e., Wnt4, Wnt5a, and Wnt11, regulate neurogenic differentiation through Fzd3 activation and the increase in downstream targets of JNK, which is one of the non-canonical pathways, in hBM-MSCs.
Collapse
Affiliation(s)
- Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Hyong-Ho Cho
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Jong-Seong Park
- Department of Physiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
36
|
Salehi H, Amirpour N, Niapour A, Razavi S. An Overview of Neural Differentiation Potential of Human Adipose Derived Stem Cells. Stem Cell Rev Rep 2016; 12:26-41. [PMID: 26490462 DOI: 10.1007/s12015-015-9631-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There is wide interest in application of adult stem cells due to easy to obtain with a minimal patient discomfort, capable of producing cell numbers in large quantities and their immunocompatible properties without restriction by ethical concerns. Among these stem cells, multipotent mesenchymal stem cells (MSCs) from human adipose tissue are considered as an ideal source for various regenerative medicine. In spite of mesodermal origin of human adipose-derived stem cells (hADSCs), these cells have differentiation potential toward mesodermal and non-mesodermal lineages. Up to now, several studies have shown that hADSCs can undergo transdifferentiation and produce cells outside of their lineage, especially into neural cells when they are transferred to a specific cell environment. The purpose of this literature review is to provide an overview of the existing state of knowledge of the differentiation potential of hADSCs, specifically their ability to give rise to neuronal cells. The following review discusses different protocols considered for differentiation of hADSCs to neural cells, the neural markers that are used in each procedure and possible mechanisms that are involved in this differentiation.
Collapse
|
37
|
Solís-Chagoyán H, Flores-Soto E, Reyes-García J, Valdés-Tovar M, Calixto E, Montaño LM, Benítez-King G. Voltage-Activated Calcium Channels as Functional Markers of Mature Neurons in Human Olfactory Neuroepithelial Cells: Implications for the Study of Neurodevelopment in Neuropsychiatric Disorders. Int J Mol Sci 2016; 17:ijms17060941. [PMID: 27314332 PMCID: PMC4926474 DOI: 10.3390/ijms17060941] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/02/2016] [Accepted: 06/07/2016] [Indexed: 11/16/2022] Open
Abstract
In adulthood, differentiation of precursor cells into neurons continues in several brain structures as well as in the olfactory neuroepithelium. Isolated precursors allow the study of the neurodevelopmental process in vitro. The aim of this work was to determine whether the expression of functional Voltage-Activated Ca2+ Channels (VACC) is dependent on the neurodevelopmental stage in neuronal cells obtained from the human olfactory epithelium of a single healthy donor. The presence of channel-forming proteins in Olfactory Sensory Neurons (OSN) was demonstrated by immunofluorescent labeling, and VACC functioning was assessed by microfluorometry and the patch-clamp technique. VACC were immunodetected only in OSN. Mature neurons responded to forskolin with a five-fold increase in Ca2+. By contrast, in precursor cells, a subtle response was observed. The involvement of VACC in the precursors’ response was discarded for the absence of transmembrane inward Ca2+ movement evoked by step depolarizations. Data suggest differential expression of VACC in neuronal cells depending on their developmental stage and also that the expression of these channels is acquired by OSN during maturation, to enable specialized functions such as ion movement triggered by membrane depolarization. The results support that VACC in OSN could be considered as a functional marker to study neurodevelopment.
Collapse
Affiliation(s)
- Héctor Solís-Chagoyán
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco No. 101, Col. San Lorenzo-Huipulco, Mexico City 14370, Mexico.
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico.
| | - Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico.
| | - Marcela Valdés-Tovar
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco No. 101, Col. San Lorenzo-Huipulco, Mexico City 14370, Mexico.
| | - Eduardo Calixto
- Departamento de Neurobiología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco No. 101, Col. San Lorenzo-Huipulco, Mexico City 14370, Mexico.
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico.
| | - Gloria Benítez-King
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco No. 101, Col. San Lorenzo-Huipulco, Mexico City 14370, Mexico.
| |
Collapse
|
38
|
Jang S, Cho HH, Kim SH, Lee KH, Cho YB, Park JS, Jeong HS. Transplantation of human adipose tissue-derived stem cells for repair of injured spiral ganglion neurons in deaf guinea pigs. Neural Regen Res 2016; 11:994-1000. [PMID: 27482231 PMCID: PMC4962600 DOI: 10.4103/1673-5374.184503] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2015] [Indexed: 12/30/2022] Open
Abstract
Excessive noise, ototoxic drugs, infections, autoimmune diseases, and aging can cause loss of spiral ganglion neurons, leading to permanent sensorineural hearing loss in mammals. Stem cells have been confirmed to be able to differentiate into spiral ganglion neurons. Little has been reported on adipose tissue-derived stem cells (ADSCs) for repair of injured spiral ganglion neurons. In this study, we hypothesized that transplantation of neural induced-human ADSCs (NI-hADSCs) can repair the injured spiral ganglion neurons in guinea pigs with neomycin-induced sensorineural hearing loss. NI-hADSCs were induced with culture medium containing basic fibroblast growth factor and forskolin and then injected to the injured cochleae. Guinea pigs that received injection of Hanks' balanced salt solution into the cochleae were used as controls. Hematoxylin-eosin staining showed that at 8 weeks after cell transplantation, the number of surviving spiral ganglion neurons in the cell transplantation group was significantly increased than that in the control group. Also at 8 weeks after cell transplantation, immunohistochemical staining showed that a greater number of NI-hADSCs in the spiral ganglions were detected in the cell transplantation group than in the control group, and these NI-hADSCs expressed neuronal markers neurofilament protein and microtubule-associated protein 2. Within 8 weeks after cell transplantation, the guinea pigs in the cell transplantation group had a gradually decreased auditory brainstem response threshold, while those in the control group had almost no response to 80 dB of clicks or pure tone burst. These findings suggest that a large amount of NI-hADSCs migrated to the spiral ganglions, survived for a period of time, repaired the injured spiral ganglion cells, and thereby contributed to the recovery of sensorineural hearing loss in guinea pigs.
Collapse
Affiliation(s)
- Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hyong-Ho Cho
- Department of Otolaryngology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Song-Hee Kim
- Department of Physiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yong-Bum Cho
- Department of Otolaryngology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jong-Seong Park
- Department of Physiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
39
|
Alghamdi S, Khan I, Beeravolu N, McKee C, Thibodeau B, Wilson G, Chaudhry GR. BET protein inhibitor JQ1 inhibits growth and modulates WNT signaling in mesenchymal stem cells. Stem Cell Res Ther 2016; 7:22. [PMID: 26830473 PMCID: PMC4736146 DOI: 10.1186/s13287-016-0278-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/04/2015] [Accepted: 01/11/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Efficacy and safety of anticancer drugs are traditionally studied using cancer cell lines and animal models. The thienodiazepine class of BET inhibitors, such as JQ1, has been extensively studied for the potential treatment of hematological malignancies and several small molecules belonging to this class are currently under clinical investigation. While these compounds are well known to inhibit cancer cell growth and cause apoptosis, their effects on stem cells, particularly mesenchymal stem cells (MSCs), which are important for regeneration of damaged cells and tissues, are unknown. In this study we employed umbilical cord derived MSCs as a model system to evaluate the safety of JQ1. METHODS Cord derived MSCs were treated with various doses of JQ1 and subjected to cell metabolic activity, apoptosis, and cell cycle analyses using MTT assay, Annexin-V/FITC and PI staining, and flow cytometry, respectively. The effect of JQ1 on gene expression was determined using microarray and quantitative real-time reverse transcriptase polymerase chain reaction analysis. Furthermore, protein expression of apoptotic and neuronal markers was carried out using western blot and immunostaining, respectively. RESULTS Our results showed that JQ1 inhibited cell growth and caused cell cycle arrest in G1 phase but did not induce apoptosis or senescence. JQ1 also down-regulated genes involved in self-renewal, cell cycle, DNA replication, and mitosis, which may have negative implications on the regenerative potential of MSCs. In addition, JQ1 interfered with signaling pathways by down regulating the expression of WNT, resulting in limiting the self-renewal. These results suggest that anticancer agents belonging to the thienodiazepine class of BET inhibitors should be carefully evaluated before their use in cancer therapy. CONCLUSIONS This study revealed for the first time that JQ1 adversely affected MSCs, which are important for repair and regeneration. JQ1 specifically modulated signal transduction and inhibited growth as well as self-renewal. These findings suggest that perinatal MSCs could be used to supplement animal models for investigating the safety of anticancer agents and other drugs.
Collapse
Affiliation(s)
- Saeed Alghamdi
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA. .,OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| | - Irfan Khan
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA. .,OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| | - Naimisha Beeravolu
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA. .,OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| | - Christina McKee
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA. .,OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| | | | - George Wilson
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA. .,Beaumont Health System, Royal Oak, MI, 48073, USA.
| | - G Rasul Chaudhry
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA. .,OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| |
Collapse
|
40
|
Unahabhokha T, Chanvorachote P, Pongrakhananon V. The attenuation of epithelial to mesenchymal transition and induction of anoikis by gigantol in human lung cancer H460 cells. Tumour Biol 2016; 37:8633-41. [PMID: 26733180 DOI: 10.1007/s13277-015-4717-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 12/21/2015] [Indexed: 12/21/2022] Open
Abstract
Lung cancer has been the major cause of death within patients due to the high metastatic rate. One of the most essential processes of metastasis is the ability of cancer cells to resist the programmed cell death in a detached condition called anoikis. The discoveries of new natural compound that is able to sensitize anoikis in cancer cells have garnered the most interest in cancer pharmaceutical science. Gigantol, a bibenzyl compound extracted from Dendrobium draconis, has been a promising natural derived compound for cancer therapy due to several cytotoxic effects in cancer cells. This study has demonstrated for the first time that gigantol significantly decreases lung cancer cells' viability in a detached condition through anoikis and anchorage-independent assays. Western blotting analysis reveals that gigantol greatly decreases epithelial to mesenchymal transition (EMT) markers including N-cadherin, vimentin, and Slug leading to a significant suppression of protein kinase B (AKT), extracellular signal-regulated kinase (ERK), and caveolin-1 (cav-1) survival pathways during the detached condition. Therefore, gigantol could be a potential cancer therapeutic compound suggesting for further development for cancer therapy.
Collapse
Affiliation(s)
- Thitita Unahabhokha
- Pharmaceutical Technology (International) Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand, 10330
- Cell-Based Drug and Health Product Development Research Unit, Chulalongkorn University, Bangkok, Thailand, 10330
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand, 10330
- Cell-Based Drug and Health Product Development Research Unit, Chulalongkorn University, Bangkok, Thailand, 10330
| | - Varisa Pongrakhananon
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand, 10330.
- Cell-Based Drug and Health Product Development Research Unit, Chulalongkorn University, Bangkok, Thailand, 10330.
| |
Collapse
|
41
|
Visweswaran M, Pohl S, Arfuso F, Newsholme P, Dilley R, Pervaiz S, Dharmarajan A. Multi-lineage differentiation of mesenchymal stem cells - To Wnt, or not Wnt. Int J Biochem Cell Biol 2015; 68:139-47. [PMID: 26410622 DOI: 10.1016/j.biocel.2015.09.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/21/2015] [Accepted: 09/22/2015] [Indexed: 01/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent precursor cells originating from several adult connective tissues. MSCs possess the ability to self-renew and differentiate into several lineages, and are recognized by the expression of unique cell surface markers. Several lines of evidence suggest that various signal transduction pathways and their interplay regulate MSC differentiation. To that end, a critical player in regulating MSC differentiation is a group of proteins encoded by the Wnt gene family, which was previously known for influencing various stages of embryonic development and cell fate determination. As MSCs have gained significant clinical attention for their potential applications in regenerative medicine, it is imperative to unravel the mechanisms by which molecular regulators control differentiation of MSCs for designing cell-based therapeutics. It is rather coincidental that the functional outcome(s) of Wnt-induced signals share similarities with cellular redox-mediated networks from the standpoint of MSC biology. Furthermore, there is evidence for a crosstalk between Wnt and redox signalling, which begs the question whether Wnt-mediated differentiation signals involve the intermediary role of reactive oxygen species. In this review, we summarize the impact of Wnt signalling on multi-lineage differentiation of MSCs, and attempt to unravel the intricate interplay between Wnt and redox signals.
Collapse
Affiliation(s)
- Malini Visweswaran
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Sebastian Pohl
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Philip Newsholme
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Rodney Dilley
- Ear Sciences Centre, University of Western Australia and Ear Science Institute Australia, Perth, Western Australia 6008, Australia
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Arun Dharmarajan
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia.
| |
Collapse
|