1
|
Liu Y, Zhao C, Zhang R, Pang Y, Li L, Feng S. Progression of mesenchymal stem cell regulation on imbalanced microenvironment after spinal cord injury. Stem Cell Res Ther 2024; 15:343. [PMID: 39354635 PMCID: PMC11446099 DOI: 10.1186/s13287-024-03914-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/01/2024] [Indexed: 10/03/2024] Open
Abstract
Spinal cord injury (SCI) results in significant neural damage and inhibition of axonal regeneration due to an imbalanced microenvironment. Extensive evidence supports the efficacy of mesenchymal stem cell (MSC) transplantation as a therapeutic approach for SCI. This review aims to present an overview of MSC regulation on the imbalanced microenvironment following SCI, specifically focusing on inflammation, neurotrophy and axonal regeneration. The application, limitations and future prospects of MSC transplantation are discussed as well. Generally, a comprehensive perspective is provided for the clinical translation of MSC transplantation for SCI.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Chenxi Zhao
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Rong Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Yilin Pang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Linquan Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Shiqing Feng
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China.
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China.
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
2
|
Adami R, Pezzotta M, Cadile F, Cuniolo B, Rovati G, Canepari M, Bottai D. Physiological Features of the Neural Stem Cells Obtained from an Animal Model of Spinal Muscular Atrophy and Their Response to Antioxidant Curcumin. Int J Mol Sci 2024; 25:8364. [PMID: 39125934 PMCID: PMC11313061 DOI: 10.3390/ijms25158364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
The most prevalent rare genetic disease affecting young individuals is spinal muscular atrophy (SMA), which is caused by a loss-of-function mutation in the telomeric gene survival motor neuron (SMN) 1. The high heterogeneity of the SMA pathophysiology is determined by the number of copies of SMN2, a separate centromeric gene that can transcribe for the same protein, although it is expressed at a slower rate. SMA affects motor neurons. However, a variety of different tissues and organs may also be affected depending on the severity of the condition. Novel pharmacological treatments, such as Spinraza, Onasemnogene abeparvovec-xioi, and Evrysdi, are considered to be disease modifiers because their use can change the phenotypes of the patients. Since oxidative stress has been reported in SMA-affected cells, we studied the impact of antioxidant therapy on neural stem cells (NSCs) that have the potential to differentiate into motor neurons. Antioxidants can act through various pathways; for example, some of them exert their function through nuclear factor (erythroid-derived 2)-like 2 (NRF2). We found that curcumin is able to induce positive effects in healthy and SMA-affected NSCs by activating the nuclear translocation of NRF2, which may use a different mechanism than canonical redox regulation through the antioxidant-response elements and the production of antioxidant molecules.
Collapse
Affiliation(s)
- Raffaella Adami
- Section of Pharmacology and Biosciences, Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (R.A.); (M.P.); (B.C.); (G.R.)
| | - Matteo Pezzotta
- Section of Pharmacology and Biosciences, Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (R.A.); (M.P.); (B.C.); (G.R.)
| | - Francesca Cadile
- Human Physiology Unit, Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy; (F.C.); (M.C.)
| | - Beatrice Cuniolo
- Section of Pharmacology and Biosciences, Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (R.A.); (M.P.); (B.C.); (G.R.)
| | - Gianenrico Rovati
- Section of Pharmacology and Biosciences, Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (R.A.); (M.P.); (B.C.); (G.R.)
| | - Monica Canepari
- Human Physiology Unit, Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy; (F.C.); (M.C.)
| | - Daniele Bottai
- Section of Pharmacology and Biosciences, Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (R.A.); (M.P.); (B.C.); (G.R.)
| |
Collapse
|
3
|
Guo X, Jiang C, Chen Z, Wang X, Hong F, Hao D. Regulation of the JAK/STAT signaling pathway in spinal cord injury: an updated review. Front Immunol 2023; 14:1276445. [PMID: 38022526 PMCID: PMC10663250 DOI: 10.3389/fimmu.2023.1276445] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Cytokines are involved in neural homeostasis and pathological processes associated with neuroinflammation after spinal cord injury (SCI). The biological effect of cytokines, including those associated with acute or chronic SCI pathologies, are the result of receptor-mediated signaling through the Janus kinases (JAKs) as well as the signal transducers and activators of transcription (STAT) DNA-binding protein families. Although therapies targeting at cytokines have led to significant changes in the treatment of SCI, they present difficulties in various aspects for the direct use by patients themselves. Several small-molecule inhibitors of JAKs, which may affect multiple pro-inflammatory cytokine-dependent pathways, as well as STATs, are in clinical development for the treatment of SCI. This review describes the current understanding of the JAK-STAT signaling in neuroendocrine homeostasis and diseases, together with the rationale for targeting at this pathway for the treatment of SCI.
Collapse
Affiliation(s)
- Xinyu Guo
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi’an, China
| | - Chao Jiang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi’an, China
| | - Zhe Chen
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi’an, China
| | - Xiaohui Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi’an, China
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Fan Hong
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi’an, China
| | - Dingjun Hao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi’an, China
| |
Collapse
|
4
|
Progression in translational research on spinal cord injury based on microenvironment imbalance. Bone Res 2022; 10:35. [PMID: 35396505 PMCID: PMC8993811 DOI: 10.1038/s41413-022-00199-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 11/14/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) leads to loss of motor and sensory function below the injury level and imposes a considerable burden on patients, families, and society. Repair of the injured spinal cord has been recognized as a global medical challenge for many years. Significant progress has been made in research on the pathological mechanism of spinal cord injury. In particular, with the development of gene regulation, cell sequencing, and cell tracing technologies, in-depth explorations of the SCI microenvironment have become more feasible. However, translational studies related to repair of the injured spinal cord have not yielded significant results. This review summarizes the latest research progress on two aspects of SCI pathology: intraneuronal microenvironment imbalance and regenerative microenvironment imbalance. We also review repair strategies for the injured spinal cord based on microenvironment imbalance, including medications, cell transplantation, exosomes, tissue engineering, cell reprogramming, and rehabilitation. The current state of translational research on SCI and future directions are also discussed. The development of a combined, precise, and multitemporal strategy for repairing the injured spinal cord is a potential future direction.
Collapse
|
5
|
Adami R, Bottai D. Spinal Muscular Atrophy Modeling and Treatment Advances by Induced Pluripotent Stem Cells Studies. Stem Cell Rev Rep 2020; 15:795-813. [PMID: 31863335 DOI: 10.1007/s12015-019-09910-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Spinal Muscular Atrophy (SMA) is a neurodegenerative disease characterized by specific and predominantly lower motor neuron (MN) loss. SMA is the main reason for infant death, while about one in 40 children born is a healthy carrier. SMA is caused by decreased levels of production of a ubiquitously expressed gene: the survival motor neuron (SMN). All SMA patients present mutations of the telomeric SMN1 gene, but many copies of a centromeric, partially functional paralog gene, SMN2, can somewhat compensate for the SMN1 deficiency, scaling inversely with phenotypic harshness. Because the study of neural tissue in and from patients presents too many challenges and is very often not feasible; the use of animal models, such as the mouse, had a pivotal impact in our understanding of SMA pathology but could not portray totally satisfactorily the elaborate regulatory mechanisms that are present in higher animals, particularly in humans. And while recent therapeutic achievements have been substantial, especially for very young infants, some issues should be considered for the treatment of older patients. An alternative way to study SMA, and other neurological pathologies, is the use of induced pluripotent stem cells (iPSCs) derived from patients. In this work, we will present a wide analysis of the uses of iPSCs in SMA pathology, starting from basic science to their possible roles as therapeutic tools.
Collapse
Affiliation(s)
- Raffaella Adami
- Department of Health Sciences, University of Milan, via A. di Rudinì 8, 20142, Milan, Italy
| | - Daniele Bottai
- Department of Health Sciences, University of Milan, via A. di Rudinì 8, 20142, Milan, Italy.
| |
Collapse
|
6
|
Papa S, Pizzetti F, Perale G, Veglianese P, Rossi F. Regenerative medicine for spinal cord injury: focus on stem cells and biomaterials. Expert Opin Biol Ther 2020; 20:1203-1213. [PMID: 32421405 DOI: 10.1080/14712598.2020.1770725] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Spinal cord injury (SCI) is a dramatic medical pathology consequence of a trauma (primary injury). However, most of the post-traumatic degeneration of the tissue is caused by the so-called secondary injury, which is known to be a multifactorial process. This, indeed, includes a wide spectrum of events: blood-brain barrier dysfunction, local inflammation, neuronal death, demyelination and disconnection of nerve pathways. AREAS COVERED Cell therapy represents a promising cure to target diseases and disorders at the cellular level, by restoring cell population or using cells as carriers of therapeutic cargo. In particular, regenerative medicine with stem cells represents the most appealing category to be used, thanks to their peculiar features. EXPERT OPINION Many preclinical research studies demonstrated that cell treatment can improve animal sensory/motor functions and so demonstrated to be very promising for clinical trials. In particular, recent advances have led to the development of biomaterials aiming to promote in situ cell delivery. This review digs into this topic discussing the possibility of cell treatment to improve medical chances in SCI repair.
Collapse
Affiliation(s)
- Simonetta Papa
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy
| | - Fabio Pizzetti
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy.,Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Milan, Italy
| | - Giuseppe Perale
- Faculty of Biomedical Sciences, University of Southern Switzerland (USI) , Lugano, Switzerland.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology , Vienna, Austria
| | - Pietro Veglianese
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Milan, Italy
| |
Collapse
|
7
|
Abstract
Objectives: The beneficial effects of many substances have been discovered because of regular dietary consumption. This is also the case with curcumin, whose effects have been known for more than 4,000 years in Eastern countries such as China and India. A curcumin-rich diet has been known to counteract many human diseases, including cancer and diabetes, and has been shown to reduce inflammation. The effect of a curcumin treatment for neurological diseases, such as spinal muscular atrophy; Alzheimer's disease; Parkinson's disease; amyotrophic lateral sclerosis; multiple sclerosis; and others, has only recently been brought to the attention of researchers and the wider population.Methods: In this paper, we summarise the studies on this natural product, from its isolation two centuries ago to its characterisation a century later.Results: We describe its role in the treatment of neurological diseases, including its cellular and common molecular mechanisms, and we report on the clinical trials of curcumin with healthy people and patients.Discussion: Commenting on the different approaches adopted by the efforts made to increase its bioavailability.
Collapse
Affiliation(s)
- Raffaella Adami
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Daniele Bottai
- Department of Health Sciences, University of Milan, Milan, Italy
| |
Collapse
|
8
|
Andreotti JP, Silva WN, Costa AC, Picoli CC, Bitencourt FCO, Coimbra-Campos LMC, Resende RR, Magno LAV, Romano-Silva MA, Mintz A, Birbrair A. Neural stem cell niche heterogeneity. Semin Cell Dev Biol 2019; 95:42-53. [PMID: 30639325 PMCID: PMC6710163 DOI: 10.1016/j.semcdb.2019.01.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/02/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022]
Abstract
In mammals, new neurons can be generated from neural stem cells in specific regions of the adult brain. Neural stem cells are characterized by their abilities to differentiate into all neural lineages and to self-renew. The specific microenvironments regulating neural stem cells, commonly referred to as neurogenic niches, comprise multiple cell populations whose precise contributions are under active current exploration. Understanding the cross-talk between neural stem cells and their niche components is essential for the development of therapies against neurological disorders in which neural stem cells function is altered. In this review, we describe and discuss recent studies that identified novel components in the neural stem cell niche. These discoveries bring new concepts to the field. Here, we evaluate these recent advances that change our understanding of the neural stem cell niche heterogeneity and its influence on neural stem cell function.
Collapse
Affiliation(s)
- Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Walison N Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alinne C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flávia C O Bitencourt
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiz A V Magno
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marco A Romano-Silva
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
9
|
Benmelouka A, Shamseldin LS, Nourelden AZ, Negida A. A Review on the Etiology and Management of Pediatric Traumatic Spinal Cord Injuries. ADVANCED JOURNAL OF EMERGENCY MEDICINE 2019; 4:e28. [PMID: 32322796 PMCID: PMC7163256 DOI: 10.22114/ajem.v0i0.256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
CONTEXT Pediatric traumatic spinal cord injury (SCI) is an uncommon presentation in the emergency department. Severe injuries are associated with devastating outcomes and complications, resulting in high costs to both the society and the economic system. EVIDENCE ACQUISITION The data on pediatric traumatic spinal cord injuries has been narratively reviewed. RESULTS Pediatric SCI is a life-threatening emergency leading to serious outcomes and high mortality in children if not managed promptly. Pediatric SCI can impose many challenges to neurosurgeons and caregivers because of the lack of large studies with high evidence level and specific guidelines in terms of diagnosis, initial management and of in-hospital treatment options. Several novel potential treatment options for SCI have been developed and are currently under investigation. However, research studies into this field have been limited by the ethical and methodological challenges. CONCLUSION Future research is needed to investigate the safety and efficacy of the recent uprising neurodegenerative techniques in SCI population. Owing to the current limitations, there is a need to develop novel trial methodologies that can overcome the current methodological and ethical limitations.
Collapse
Affiliation(s)
| | | | | | - Ahmed Negida
- Medical Research Group of Egypt, Egypt
- Faculty of Medicine, Zagazig University, Zagazig, Egypt
- Neurosurgery Department, Bahçeşehir University, Istanbul, Turkey
| |
Collapse
|
10
|
Shao A, Tu S, Lu J, Zhang J. Crosstalk between stem cell and spinal cord injury: pathophysiology and treatment strategies. Stem Cell Res Ther 2019; 10:238. [PMID: 31387621 PMCID: PMC6683526 DOI: 10.1186/s13287-019-1357-z] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The injured spinal cord is difficult to repair and regenerate. Traditional treatments are not effective. Stem cells are a type of cells that have the potential to differentiate into various cells, including neurons. They exert a therapeutic effect by safely and effectively differentiating into neurons or replacing damaged cells, secreting neurotrophic factors, and inhibiting the inflammatory response. Many types of stem cells have been used for transplantation, and each has its own advantages and disadvantages. This review discusses the possible mechanisms of stem cell therapy for spinal cord injury, and the types of stem cells commonly used in experiments, to provide a reference for basic and clinical research on stem cell therapy for spinal cord injury.
Collapse
Affiliation(s)
- Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| | - Sheng Tu
- Department of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jianan Lu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.,Brain Research Institute, Zhejiang University, Hangzhou, 310003, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, 310003, China
| |
Collapse
|
11
|
Cofano F, Boido M, Monticelli M, Zenga F, Ducati A, Vercelli A, Garbossa D. Mesenchymal Stem Cells for Spinal Cord Injury: Current Options, Limitations, and Future of Cell Therapy. Int J Mol Sci 2019; 20:ijms20112698. [PMID: 31159345 PMCID: PMC6600381 DOI: 10.3390/ijms20112698] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) constitutes an inestimable public health issue. The most crucial phase in the pathophysiological process of SCI concerns the well-known secondary injury, which is the uncontrolled and destructive cascade occurring later with aberrant molecular signaling, inflammation, vascular changes, and secondary cellular dysfunctions. The use of mesenchymal stem cells (MSCs) represents one of the most important and promising tested strategies. Their appeal, among the other sources and types of stem cells, increased because of their ease of isolation/preservation and their properties. Nevertheless, encouraging promise from preclinical studies was followed by weak and conflicting results in clinical trials. In this review, the therapeutic role of MSCs is discussed, together with their properties, application, limitations, and future perspectives.
Collapse
Affiliation(s)
- Fabio Cofano
- Department of Neuroscience "Rita Levi Montalcini", Neurosurgery Unit, University of Turin, 10126 Turin, Italy.
| | - Marina Boido
- Department of Neuroscience "Rita Levi Montalcini", Neuroscience Institute "Cavalieri Ottolenghi", University of Turin, Consorzio Istituto Nazionale di Neuroscienze, 10043 Orbassano, Italy.
| | - Matteo Monticelli
- Department of Neuroscience "Rita Levi Montalcini", Neurosurgery Unit, University of Turin, 10126 Turin, Italy.
| | - Francesco Zenga
- Department of Neuroscience "Rita Levi Montalcini", Neurosurgery Unit, University of Turin, 10126 Turin, Italy.
| | - Alessandro Ducati
- Department of Neuroscience "Rita Levi Montalcini", Neurosurgery Unit, University of Turin, 10126 Turin, Italy.
| | - Alessandro Vercelli
- Department of Neuroscience "Rita Levi Montalcini", Neuroscience Institute "Cavalieri Ottolenghi", University of Turin, Consorzio Istituto Nazionale di Neuroscienze, 10043 Orbassano, Italy.
| | - Diego Garbossa
- Department of Neuroscience "Rita Levi Montalcini", Neurosurgery Unit, University of Turin, 10126 Turin, Italy.
| |
Collapse
|
12
|
Stem cell paracrine effect and delivery strategies for spinal cord injury regeneration. J Control Release 2019; 300:141-153. [PMID: 30851286 DOI: 10.1016/j.jconrel.2019.02.038] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/29/2022]
Abstract
Spinal cord injury (SCI) is a complicated neuropathological condition that results in functional dysfunction and paralysis. Various treatments have been proposed including drugs, biological factors and cells administered in several ways. Stem cell therapy offers a potentially revolutionary mode to repair the damaged spinal cord after injury. Initially, stem cells were considered promising for replacing cells and tissue lost after SCI. Many studies looked at their differentiation to replace neuronal and glial cells for a better functional outcome. However, it is becoming clear that different functional improvements recognized to stem cells are due to biomolecular activities by the transplanted stem cells rather than cell replacement. This review aimed to discuss the paracrine mechanisms for tissue repair and regeneration after stem cell transplantation in SCI. It focuses on stem cell factor production, effect in tissue restoration, and novel delivery strategies to use them for SCI therapy.
Collapse
|
13
|
Bottai D, Spreafico M, Pistocchi A, Fazio G, Adami R, Grazioli P, Canu A, Bragato C, Rigamonti S, Parodi C, Cazzaniga G, Biondi A, Cotelli F, Selicorni A, Massa V. Modeling Cornelia de Lange syndrome in vitro and in vivo reveals a role for cohesin complex in neuronal survival and differentiation. Hum Mol Genet 2019; 28:64-73. [PMID: 30239720 DOI: 10.1093/hmg/ddy329] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/12/2018] [Indexed: 02/07/2023] Open
Abstract
Cornelia de Lange syndrome (CdLS), which is reported to affect ∼1 in 10 000 to 30 000 newborns, is a multisystem organ developmental disorder with relatively mild to severe effects. Among others, intellectual disability represents an important feature of this condition. CdLS can result from mutations in at least five genes: nipped-B-like protein, structural maintenance of chromosomes 1A, structural maintenance of chromosomes 3, RAD21 cohesin complex component and histone deacetylase 8 (HDAC8). It is believed that mutations in these genes cause CdLS by impairing the function of the cohesin complex (to which all the aforementioned genes contribute to the structure or function), disrupting gene regulation during critical stages of early development. Since intellectual disorder might result from alterations in neural development, in this work, we studied the role of Hdac8 gene in mouse neural stem cells (NSCs) and in vertebrate (Danio rerio) brain development by knockdown and chemical inhibition experiments. Underlying features of Hdac8 deficiency is an increased cell death in the developing neural tissues, either in mouse NSCs or in zebrafish embryos.
Collapse
Affiliation(s)
- Daniele Bottai
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| | - Marco Spreafico
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Anna Pistocchi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Grazia Fazio
- Centro Ricerca Tettamanti, Clinica Pediatrica, Università degli Studi di Milano-Bicocca, Fondazione MBBM/Ospedale S. Gerardo, Monza, Italy
| | - Raffaella Adami
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| | - Paolo Grazioli
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| | - Adriana Canu
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Cinzia Bragato
- Fondazione IRCCS Istituto Neurologico C. Besta, Milano, Italy
- PhD program in Neuroscience, University of Milano-Bicocca, Monza, Italy
| | - Silvia Rigamonti
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
- Centro Ricerca Tettamanti, Clinica Pediatrica, Università degli Studi di Milano-Bicocca, Fondazione MBBM/Ospedale S. Gerardo, Monza, Italy
| | - Chiara Parodi
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| | - Gianni Cazzaniga
- Centro Ricerca Tettamanti, Clinica Pediatrica, Università degli Studi di Milano-Bicocca, Fondazione MBBM/Ospedale S. Gerardo, Monza, Italy
| | - Andrea Biondi
- Clinica Pediatrica, Università degli Studi di Milano-Bicocca, Fondazione MBBM/Ospedale S. Gerardo, Monza, Italy
| | - Franco Cotelli
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| | | | - Valentina Massa
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
14
|
Tamburini E, Dallatomasina A, Quartararo J, Cortelazzi B, Mangieri D, Lazzaretti M, Perris R. Structural deciphering of the NG2/CSPG4 proteoglycan multifunctionality. FASEB J 2018; 33:3112-3128. [PMID: 30550356 DOI: 10.1096/fj.201801670r] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The chondroitin sulfate proteoglycan 4 ( CSPG4) gene encodes a transmembrane proteoglycan (PG) constituting the largest and most structurally complex macromolecule of the human surfaceome. Its transcript shows an extensive evolutionary conservation and, due to the elaborated intracellular processing of the translated protein, it generates an array of glycoforms with the potential to exert variant-specific functions. CSPG4-mediated molecular events are articulated through the interaction with more than 40 putative ligands and the concurrent involvement of the ectodomain and cytoplasmic tail. Alternating inside-out and outside-in signal transductions may thereby be elicited through a tight functional connection of the PG with the cytoskeleton and its regulators. The potential of CSPG4 to influence both types of signaling mechanisms is also asserted by its lateral mobility along the plasma membrane and its intersection with microdomain-restricted internalization and endocytic trafficking. Owing to the multitude of molecular interplays that CSPG4 may engage, and thanks to a differential phosphorylation of its intracellular domain accounted by crosstalking signaling pathways, the PG stands out for its unique capability to affect numerous cellular phenomena, including those purporting pathologic conditions. We discuss here the progresses made in advancing our understanding about the structural-functional bases for the ability of CSPG4 to widely impact on cell behavior, such as to highlight how its multivalency may be exploited to interfere with disease progression.-Tamburini, E., Dallatomasina, A., Quartararo, J., Cortelazzi, B., Mangieri, D., Lazzaretti, M., Perris, R. Structural deciphering of the NG2/CSPG4 proteoglycan multifunctionality.
Collapse
Affiliation(s)
- Elisa Tamburini
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | - Alice Dallatomasina
- Division of Experimental Oncology, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy; and
| | - Jade Quartararo
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | - Barbara Cortelazzi
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | | | - Mirca Lazzaretti
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | - Roberto Perris
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| |
Collapse
|
15
|
Bottai D, Adami R, Ghidoni R. The crosstalk between glycosphingolipids and neural stem cells. J Neurochem 2018; 148:698-711. [PMID: 30269334 DOI: 10.1111/jnc.14600] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 09/13/2018] [Accepted: 09/18/2018] [Indexed: 01/19/2023]
Abstract
Until a few years ago, the majority of cell functions were envisioned as the result of protein and DNA activity. The cell membranes were considered as a mere structure of support and/or separation. In the last years, the function of cell membranes has, however, received more attention and their components of lipid nature have also been depicted as important cell mediators and the membrane organization was described as an important determinant for membrane-anchored proteins activity. In particular, because of their high diversity, glycosphingolipids offer a wide possibility of regulation. Specifically, the role of glycosphingolipids, in the fine-tuning of neuron activity, has recently received deep attention. For their pivotal role in vertebrate and mammals neural development, neural stem cells regulation is of main interest especially concerning their further functions in neurological pathology progression and treatment. Glycosphingolipids expression present a developmental regulation. In this view, glycosphingolipids can hold an important role in neural stem cells features because of their heterogeneity and their consequent capacity for eclectic interaction with other cell components.
Collapse
Affiliation(s)
- Daniele Bottai
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Raffaella Adami
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Riccardo Ghidoni
- Department of Health Sciences, University of Milan, Milan, Italy
| |
Collapse
|
16
|
Adami R, Pagano J, Colombo M, Platonova N, Recchia D, Chiaramonte R, Bottinelli R, Canepari M, Bottai D. Reduction of Movement in Neurological Diseases: Effects on Neural Stem Cells Characteristics. Front Neurosci 2018; 12:336. [PMID: 29875623 PMCID: PMC5974544 DOI: 10.3389/fnins.2018.00336] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/30/2018] [Indexed: 01/04/2023] Open
Abstract
Both astronauts and patients affected by chronic movement-limiting pathologies face impairment in muscle and/or brain performance. Increased patient survival expectations and the expected longer stays in space by astronauts may result in prolonged motor deprivation and consequent pathological effects. Severe movement limitation can influence not only the motor and metabolic systems but also the nervous system, altering neurogenesis and the interaction between motoneurons and muscle cells. Little information is yet available about the effect of prolonged muscle disuse on neural stem cells characteristics. Our in vitro study aims to fill this gap by focusing on the biological and molecular properties of neural stem cells (NSCs). Our analysis shows that NSCs derived from the SVZ of HU mice had shown a reduced proliferation capability and an altered cell cycle. Furthermore, NSCs obtained from HU animals present an incomplete differentiation/maturation. The overall results support the existence of a link between reduction of exercise and muscle disuse and metabolism in the brain and thus represent valuable new information that could clarify how circumstances such as the absence of load and the lack of movement that occurs in people with some neurological diseases, may affect the properties of NSCs and contribute to the negative manifestations of these conditions.
Collapse
Affiliation(s)
- Raffaella Adami
- Department of Health Science, University of Milan, Milan, Italy
| | - Jessica Pagano
- Department of Health Science, University of Milan, Milan, Italy
| | - Michela Colombo
- Department of Health Science, University of Milan, Milan, Italy
| | | | - Deborah Recchia
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | | | - Monica Canepari
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Daniele Bottai
- Department of Health Science, University of Milan, Milan, Italy
| |
Collapse
|
17
|
Jensen TJ, Shui JE, Finck CM. The effect of meconium exposure on the expression and differentiation of amniotic fluid mesenchymal stem cells. J Neonatal Perinatal Med 2018; 10:313-323. [PMID: 28854517 PMCID: PMC5676976 DOI: 10.3233/npm-16141] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND: The goal of this study was to determine if exposure to meconium would alter the phenotype of amniotic fluid mesenchymal stem cells (AF-MSCs) and the ability of these cells to be differentiated into distal airway type cells. METHODS: Meconium was collected, lyophilized and resuspended in PBS at 3 different concentrations (high, medium, and low). AF-MSCs were cultured in the presence of this meconium suspension for 8 hours and then analyzed for changes in gene expression. Additionally, AF-MSCs exposed to meconium were differentiated for 14 days using modified small airway growth medium (mSAGM) and gene expression was determined. As a spontaneous differentiation control, meconium exposed AF-MSCs were cultured in amniotic fluid stem cell medium (AF medium). RESULTS: After 8 hours of exposure in culture, AF-MSCs had increased expression of distal airway genes aquaporin 5 (AQP5) and surfactant protein c (SPC) when cultured in AF medium containing meconium. These gene expression levels were similar to that of AF-MSCs that were differentiated in mSAGM for 14 days. Furthermore, there was an up regulation of pluripotency genes NANOG and OCT4 in response to low meconium concentration for 8 hours. Following 14 days of culture in mSAGM, there was an upregulation of TTF1, SPC and AQP5 expression in the control, as well as in the low and medium meconium exposed groups indicating that these cells were still able to be differentiated. High meconium concentration did, however, appear to influence the level of distal airway gene expression after 14 days in mSAGM. After 14 days in AF medium, there was significant downregulation in pluripotency and mesenchymal markers as well as distal airway gene expression in all groups. CONCLUSION: The phenotype of AF-MSCs is modulated by meconium exposure; however, the cells were still able to differentiate into distal airway gene and protein expression. This result supports the hypothesis that progenitor cells exist in the amniotic fluid and the presence of meconium may affect their initial phenotype. However, these cells were still able to be differentiated to a distal lung phenotype.
Collapse
Affiliation(s)
- T J Jensen
- Department of Pediatrics, UConn Health, Farmington, CT, USA
| | - J E Shui
- Department of Pediatrics, Connecticut Children's Medical Center, Hartford, CT, USA
| | - C M Finck
- Department of Surgery, Connecticut Children's Medical Center, Harford, CT, USA
| |
Collapse
|
18
|
Vismara I, Papa S, Rossi F, Forloni G, Veglianese P. Current Options for Cell Therapy in Spinal Cord Injury. Trends Mol Med 2017; 23:831-849. [PMID: 28811172 DOI: 10.1016/j.molmed.2017.07.005] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/13/2017] [Accepted: 07/16/2017] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) is a complex pathology that evolves after primary acute mechanical injury, causing further damage to the spinal cord tissue that exacerbates clinical outcomes. Based on encouraging results from preclinical experiments, some cell treatments being translated into clinical practice demonstrate promising and effective improvement in sensory/motor function. Combinatorial treatments of cell and drug/biological factors have been demonstrated to be more effective than cell treatments alone. Recent advances have led to the development of biomaterials aiming to promote in situ cell delivery for SCI, together with combinatorial strategies using drugs/biomolecules to achieve a maximized multitarget approach. This review provides an overview of single and combinatorial regenerative cell treatments as well as potential delivery options to treat SCI.
Collapse
Affiliation(s)
- Irma Vismara
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy; These authors contributed equally to this work
| | - Simonetta Papa
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy; These authors contributed equally to this work
| | - Filippo Rossi
- Dipartimento di Chimica, Materiali e Ingegneria Chimica 'Giulio Natta', Politecnico di Milano, via Mancinelli 7, 20131 Milano, Italy
| | - Gianluigi Forloni
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy
| | - Pietro Veglianese
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy.
| |
Collapse
|
19
|
Hamid AA, Joharry MK, Mun-Fun H, Hamzah SN, Rejali Z, Yazid MN, Thilakavathy K, Nordin N. Highly potent stem cells from full-term amniotic fluid: A realistic perspective. Reprod Biol 2017; 17:9-18. [PMID: 28262444 DOI: 10.1016/j.repbio.2017.02.001] [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: 03/31/2016] [Revised: 01/31/2017] [Accepted: 02/10/2017] [Indexed: 12/19/2022]
Abstract
Amniotic fluid (AF) is now known to harbor highly potent stem cells, making it an excellent source for cell therapy. However, most of the stem cells isolated are from AF of mid-term pregnancies in which the collection procedure involves an invasive technique termed amniocentesis. This has limited the access in getting the fluid as the technique imposes certain level of risks to the mother as well as to the fetus. Alternatively, getting AF from full-term pregnancies or during deliveries would be a better resolution. Unfortunately, very few studies have isolated stem cells from AF at this stage of gestation, the fluid that is merely discarded. The question remains whether full-term AF harbors stem cells of similar potency as of the stem cells of mid-term AF. Here, we aim to review the prospect of having this type of stem cells by first looking at the origin and contents of AF particularly during different gestation period. We will then discuss the possibility that the AF, at full term, contains a population of highly potent stem cells. These stem cells are distinct from, and probably more potent than the AF mesenchymal stem cells (AF-MSCs) isolated from full-term AF. By comparing the studies on stem cells isolated from mid-term versus full-term AF from various species, we intend to address the prospect of having highly potent amniotic fluid stem cells from AF of full-term pregnancies in human and animals.
Collapse
Affiliation(s)
- Adila A Hamid
- Stem Cell Research Laboratory, Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia; Genetics & Regenerative Medicine Research Centre, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia; Department of Physiology, Faculty of Medicine, National University of Malaysia Medical Centre, Kuala Lumpur, Malaysia.
| | - Muhammad Khair Joharry
- Stem Cell Research Laboratory, Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia; Genetics & Regenerative Medicine Research Centre, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia.
| | - Hoo Mun-Fun
- Stem Cell Research Laboratory, Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia; Genetics & Regenerative Medicine Research Centre, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia.
| | - Siti Nurusaadah Hamzah
- Stem Cell Research Laboratory, Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia; Genetics & Regenerative Medicine Research Centre, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia.
| | - Zulida Rejali
- Department of Obstetrics and Gynaecology, Universiti Putra Malaysia, Malaysia.
| | - Mohd Nazri Yazid
- Department of Obstetrics and Gynaecology, Universiti Putra Malaysia, Malaysia.
| | - Karuppiah Thilakavathy
- Stem Cell Research Laboratory, Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia; Genetics & Regenerative Medicine Research Centre, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia.
| | - Norshariza Nordin
- Stem Cell Research Laboratory, Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia; Genetics & Regenerative Medicine Research Centre, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Malaysia.
| |
Collapse
|
20
|
Di Trapani M, Bassi G, Fontana E, Giacomello L, Pozzobon M, Guillot PV, De Coppi P, Krampera M. Immune regulatory properties of CD117(pos) amniotic fluid stem cells vary according to gestational age. Stem Cells Dev 2015; 24:132-43. [PMID: 25072397 DOI: 10.1089/scd.2014.0234] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Amniotic Fluid Stem (AFS) cells are broadly multipotent fetal stem cells derived from the positive selection and ex vivo expansion of amniotic fluid CD117/c-kit(pos) cells. Considering the differentiation potential in vitro toward cell lineages belonging to the three germ layers, AFS cells have raised great interest as a new therapeutic tool, but their immune properties still need to be assessed. We analyzed the in vitro immunological properties of AFS cells from different gestational age in coculture with T, B, and natural killer (NK) cells. Nonactivated (resting) first trimester-AFS cells showed lower expression of HLA class-I molecules and NK-activating ligands than second and third trimester-AFS cells, whose features were associated with lower sensitivity to NK cell-mediated lysis. Nevertheless, inflammatory priming with interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) enhanced resistance of all AFS cell types to NK cytotoxicity. AFS cells modulated lymphocyte proliferation in a different manner according to gestational age: first trimester-AFS cells significantly inhibited T and NK cell proliferation, while second and third trimester-AFS cells were less efficient. In addition, only inflammatory-primed second trimester-AFS cells could suppress B cell proliferation, which was not affected by the first and third trimester-AFS cells. Indolamine 2,3 dioxygenase pathway was significantly involved only in T cell suppression mediated by second and third trimester-AFS cells. Overall, this study shows a number of significant quantitative differences among AFS cells of different gestational age that have to be considered in view of their clinical application.
Collapse
Affiliation(s)
- Mariano Di Trapani
- 1 Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona , Verona, Italy
| | | | | | | | | | | | | | | |
Collapse
|
21
|
The targeted inhibitory effects of human amniotic fluid stem cells carrying CXCR4 promoter and DAL-1 on non-small cell lung carcinoma growth. Gene Ther 2015; 23:214-22. [DOI: 10.1038/gt.2015.90] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 05/11/2015] [Accepted: 08/05/2015] [Indexed: 12/16/2022]
|
22
|
Rat full term amniotic fluid harbors highly potent stem cells. Res Vet Sci 2015; 102:89-99. [PMID: 26412526 DOI: 10.1016/j.rvsc.2015.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 06/30/2015] [Accepted: 07/13/2015] [Indexed: 01/08/2023]
Abstract
Amniotic fluid stem cells (AFSCs) are commonly isolated from mid-term amniotic fluid (AF) of animals and human collected via an invasive technique, amniocentesis. Alternatively, AFSCs could be collected at full-term. However, it is unclear whether AFSCs are present in the AF at full term. Here, we aimed to isolate and characterize stem cells isolated from AF of full term pregnant rats. Three stem cell lines have been established following immuno-selection against the stem cell marker, c-kit. Two of the new lines expressed multiple markers of pluripotency until more than passage 90. Further, they spontaneously differentiated into derivatives of the three primary germ layers through the formation of good quality embryoid bodies (EBs), and can be directly differentiated into neural lineage. Their strong stemness and potent neurogenic properties highlight the presence of highly potent stem cells in AF of full-term pregnancies, which could serve as a potential source of stem cells for regenerative medicine.
Collapse
|
23
|
Liu R, Zhang S, Yang H, Ju P, Xia Y, Shi Y, Lim TH, Lim AS, Liang F, Feng Z. Characterization and therapeutic evaluation of a Nestin+ CNP+ NG2+ cell population on mouse spinal cord injury. Exp Neurol 2015; 269:28-42. [DOI: 10.1016/j.expneurol.2015.03.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 03/17/2015] [Accepted: 03/22/2015] [Indexed: 11/29/2022]
|
24
|
Maraldi T, Beretti F, Guida M, Zavatti M, De Pol A. Role of hepatocyte growth factor in the immunomodulation potential of amniotic fluid stem cells. Stem Cells Transl Med 2015; 4:539-47. [PMID: 25873747 DOI: 10.5966/sctm.2014-0266] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/23/2015] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Human amniotic fluid stem cells (hAFSCs) may be useful for regenerative medicine because of their potential to differentiate into all three germ layers and to modulate immune response with different types of secretion molecules. This last issue has not been completely elucidated. The aim of this study was to investigate the secretome profile of the hAFSC, focusing on the role of hepatocyte growth factor (HGF) in immunoregulation through short and long cocultures with human peripheral blood mononuclear cells. We found that HGF produced by hAFSCs exerts a cytoprotective role, inducing an increase in caspase-dependent apoptosis in human immune cells. This study provides evidence supporting the hypothesis that amniotic fluid is an ideal source of stem cells for expansion and banking properties for therapeutic use. hAFSCs not only are less immunogenic but also can secrete immunoregulatory factors that may be useful in autoimmune diseases or allogenic implants. SIGNIFICANCE New information about the secretome pattern is reported in this paper. Human amniotic fluid stem cells (hAFSCs) possess immunomodulatory properties involving hepatocyte growth factor production. hAFSCs could be used in immunotherapies and might be able to avoid allogenic rejection.
Collapse
Affiliation(s)
- Tullia Maraldi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Francesca Beretti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Marianna Guida
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Manuela Zavatti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Anto De Pol
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| |
Collapse
|
25
|
Adami R, Scesa G, Bottai D. Stem cell transplantation in neurological diseases: improving effectiveness in animal models. Front Cell Dev Biol 2014; 2:17. [PMID: 25364724 PMCID: PMC4206985 DOI: 10.3389/fcell.2014.00017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 04/22/2014] [Indexed: 12/14/2022] Open
Abstract
Neurological diseases afflict a growing proportion of the human population. There are two reasons for this: first, the average age of the population (especially in the industrialized world) is increasing, and second, the diagnostic tools to detect these pathologies are now more sophisticated and can be used on a higher percentage of the population. In many cases, neurological disease has a pharmacological treatment which, as in the case of Alzheimer's disease, Parkinson's disease, Epilepsy, and Multiple Sclerosis can reduce the symptoms and slow down the course of the disease but cannot reverse its effects or heal the patient. In the last two decades the transplantation approach, by means of stem cells of different origin, has been suggested for the treatment of neurological diseases. The choice of slightly different animal models and the differences in methods of stem cell preparation make it difficult to compare the results of transplantation experiments. Moreover, the translation of these results into clinical trials with human subjects is difficult and has so far met with little success. This review seeks to discuss the reasons for these difficulties by considering the differences between human and animal cells (including isolation, handling and transplantation) and between the human disease model and the animal disease model.
Collapse
Affiliation(s)
- Raffaella Adami
- Department of Health Science, Faculty of Medicine, University of Milan Milan, Italy
| | - Giuseppe Scesa
- Department of Health Science, Faculty of Medicine, University of Milan Milan, Italy
| | - Daniele Bottai
- Department of Health Science, Faculty of Medicine, University of Milan Milan, Italy
| |
Collapse
|