scholarly journals Stem cell sources and therapeutic approaches for central nervous system and neural retinal disorders

2008 ◽  
Vol 24 (3-4) ◽  
pp. E11 ◽  
Author(s):  
Diana Yu ◽  
Gabriel A. Silva

✓ In the past decades, stem cell biology has made a profound impact on our views of mammalian development as well as opened new avenues in regenerative medicine. The potential of stem cells to differentiate into various cell types of the body is the principal reason they are being explored in treatments for diseases in which there may be dysfunctional cells and/or loss of healthy cells due to disease. In addition, other properties are unique to stem cells; their endogenous trophic support, ability to home to sites of pathological entities, and stability in culture, which allows genetic manipulation, are also being utilized to formulate stem cell–based therapy for central nervous system (CNS) disorders. In this review, the authors will review key characteristics of embryonic and somatic (adult) stem cells, consider therapeutic strategies employed in stem cell therapy, and discuss the recent advances made in stem cell–based therapy for a number of progressive neurodegenerative diseases in the CNS as well as neuronal degeneration secondary to other abnormalities and injuries. Although a great deal of progress has been made in our knowledge of stem cells and their utility in treating CNS disorders, much still needs to be elucidated regarding the biology of the stem cells and the pathogenesis of targeted CNS diseases to maximize therapeutic benefits. Nonetheless, stem cells present tremendous promise in the treatment of a variety of neurodegenerative diseases.

Author(s):  
Prithiv K R Kumar

Stem cells have the capacity to differentiate into any type of cell or organ. Stems cell originate from any part of the body, including the brain. Brain cells or rather neural stem cells have the capacitive advantage of differentiating into the central nervous system leading to the formation of neurons and glial cells. Neural stem cells should have a source by editing DNA, or by mixings chemical enzymes of iPSCs. By this method, a limitless number of neuron stem cells can be obtained. Increase in supply of NSCs help in repairing glial cells which in-turn heal the central nervous system. Generally, brain injuries cause motor and sensory deficits leading to stroke. With all trials from novel therapeutic methods to enhanced rehabilitation time, the economy and quality of life is suppressed. Only PSCs have proven effective for grafting cells into NSCs. Neurons derived from stem cells is the only challenge that limits in-vitro usage in the near future.


Development ◽  
2000 ◽  
Vol 127 (6) ◽  
pp. 1277-1290 ◽  
Author(s):  
L. Mathis ◽  
J.F. Nicolas

We have performed a systematic clonal analysis to describe the modes of growth, dispersion and production of cells during the development of the mouse neural system. We have used mice expressing a LaacZ reporter gene under the control of the neuron specific enolase promoter to randomly generate LacZ clones in the central nervous system (CNS). We present evidence for (1) a pool of CNS founder cells that is not regionalized, i.e. give descendants dispersed along the entire A-P axis, (2) an early separation between pools of precursors for the anterior and posterior CNS and (3) distinct modes of production of progenitors in these two domains. More specifically, cell growth and dispersion of the progenitors follow a relatively coherent pattern throughout the anterior CNS, a mode that leads to a progressive regionalization of cell fates. In contrast, cell growth of progenitors of the SC appears to involve self-renewing stem cells that progress caudally during regression of the mode. Therefore, at least part of the area surrounding the node is composed of precursors with self-renewing properties and the development of the trunk is dependent on pools of stem cells regressing from A to P. Taken together with our analysis of the cell growth changes associated with neuromere formation (Mathis, L., Sieur, J., Voiculescu, O., Charnay, P. and Nicolas, J. F. (1999) Development 126, 4095–4106), our results suggest that major transitions in CNS development correspond to changes in cell behavior and may provide a link between morphogenesis and genetic patterning mechanisms (i.e. formation of the body plan).


2011 ◽  
Vol 2011 ◽  
pp. 1-11 ◽  
Author(s):  
Jonathan Witherick ◽  
Alastair Wilkins ◽  
Neil Scolding ◽  
Kevin Kemp

Although significant advances have recently been made in the understanding and treatment of multiple sclerosis, reduction of long-term disability remains a key goal. Evidence suggests that inflammation and oxidative stress within the central nervous system are major causes of ongoing tissue damage in the disease. Invading inflammatory cells, as well as resident central nervous system cells, release a number of reactive oxygen and nitrogen species which cause demyelination and axonal destruction, the pathological hallmarks of multiple sclerosis. Reduction in oxidative damage is an important therapeutic strategy to slow or halt disease processes. Many drugs in clinical practice or currently in trial target this mechanism. Cell-based therapies offer an alternative source of antioxidant capability. Classically thought of as being important for myelin or cell replacement in multiple sclerosis, stem cells may, however, have a more important role as providers of supporting factors or direct attenuators of the disease. In this paper we focus on the antioxidant properties of mesenchymal stem cells and discuss their potential importance as a cell-based therapy for multiple sclerosis.


Cells ◽  
2020 ◽  
Vol 9 (11) ◽  
pp. 2517
Author(s):  
Elizabeth Ford ◽  
Jodie Pearlman ◽  
Travis Ruan ◽  
John Manion ◽  
Matthew Waller ◽  
...  

Neurodegenerative diseases are characterized by irreversible cell damage, loss of neuronal cells and limited regeneration potential of the adult nervous system. Pluripotent stem cells are capable of differentiating into the multitude of cell types that compose the central and peripheral nervous systems and so have become the major focus of cell replacement therapies for the treatment of neurological disorders. Human embryonic stem cell (hESC) and human induced pluripotent stem cell (hiPSC)-derived cells have both been extensively studied as cell therapies in a wide range of neurodegenerative disease models in rodents and non-human primates, including Parkinson’s disease, stroke, epilepsy, spinal cord injury, Alzheimer’s disease, multiple sclerosis and pain. In this review, we discuss the latest progress made with stem cell therapies targeting these pathologies. We also evaluate the challenges in clinical application of human pluripotent stem cell (hPSC)-based therapies including risk of oncogenesis and tumor formation, immune rejection and difficulty in regeneration of the heterogeneous cell types composing the central nervous system.


2021 ◽  
Vol 271 ◽  
pp. 03076
Author(s):  
Weibai Chen

Neural stem cells have the ability to proliferation, differentiate and renew, which plays an important role in the growth, maturation and senescence of the human brain. But according to researches, neural stem cells in the brain do not remain active throughout an organism's lifetime. Many neural stem cells become dormant when the brain matures, and may be activated when the body is sick to selectively heal the disease. In recent years, there are many studies on neural stem cells. Joshua[1] and Ting Zhang[2] show that neurodegenerative diseases such as ischemic stroke, Alzheimer's disease and Parkinson's disease can be improved by the transplantation of neural stem cells, however the specific mechanism is not clear. This paper investigates three main questions: Why neural stem cell transplantation is chosen as a treatment? Where does NSCs derive from in clinical transplantation? How does neural stem cell transplantation treat brain diseases? And we also figure out the answers to these three questions. Firstly, transplantation of hypothalamic NSCs can delay the process of aging in the host, and Chemokines and EVs which secreted by neural stem cells can delay aging and defend neurodegenerative diseases. Secondly, the sources of NSCs can be divided into three types. The first is to isolate NSCs from primary tissue and cultivate them in vitro. The second is to produce the required cells by inducing pluripotent stem cells and embryonic stem cells. The third way to get NCS is through transdifferentiation of somatic cells. Thirdly, in brain diseases, transplanted NSCs can migrate from the aggregation site to the site of the disease, reducing damage to the blood-brain barrier, repairing learning and memory abilities that depend on the hippocampus and secreting neurotrophic factors.


2016 ◽  
Vol 27 (5) ◽  
pp. 491-499 ◽  
Author(s):  
Lei Wang ◽  
Yan Liu ◽  
Shaiqi Sun ◽  
Ming Lu ◽  
Ying Xia

AbstractNeural stem cell transplantation is becoming a promising and attractive cell-based treatment modality for repairing the damaged central nervous system. One of the limitations of this approach is that the proportion of functional cells differentiated from stem cells still remains at a low level. In recent years, novel long non-coding RNAs (lncRNAs) are being discovered at a growing pace, suggesting that this class of molecules may act as novel regulators in neuronal-glial fate specification. In this review, we first describe the general features of lncRNAs that are more likely to be relevant to reveal their function. By this, we aim to point out the specific roles of a number of lncRNAs whose function has been described during neuronal and glial cell differentiation. There is no doubt that investigation of the lncRNAs will open a new window in studying neuronal-glial fate specification.


2003 ◽  
Vol 31 (1) ◽  
pp. 45-49 ◽  
Author(s):  
M.P. Stavridis ◽  
A.G. Smith

Pluripotent embryonic stem cells can give rise to neuroectodermal derivatives in culture. This potential could be harnessed to generate neurons and glia for cell-replacement therapies in the central nervous system and for use in drug discovery. However, current methods of neural differentiation are empirical and relatively innefficient. Here, we review these methodologies and present new tools for quantification, analysis and manipulation of embryonic stem cell neural determination.


Biology ◽  
2021 ◽  
Vol 10 (3) ◽  
pp. 172
Author(s):  
Shivkanya Fuloria ◽  
Vetriselvan Subramaniyan ◽  
Rajiv Dahiya ◽  
Sunita Dahiya ◽  
Kalvatala Sudhakar ◽  
...  

Evidence suggests that stem cells exert regenerative potential via the release of extracellular vesicles. Mesenchymal stem cell extracellular vesicles (MSCEVs) offer therapeutic benefits for various pathophysiological ailments by restoring tissues. Facts suggest that MSCEV action can be potentiated by modifying the mesenchymal stem cells culturing methodology and bioengineering EVs. Limited clinical trials of MSCEVs have questioned their superiority, culturing quality, production scale-up and isolation, and administration format. Translation of preclinically successful MSCEVs into a clinical platform requires paying attention to several critical matters, such as the production technique, quantification/characterization, pharmacokinetics/targeting/transfer to the target site, and the safety profile. Keeping these issues as a priority, the present review was designed to highlight the challenges in translating preclinical MSCEV research into clinical platforms and provide evidence for the regenerative potential of MSCEVs in various conditions of the liver, kidney, heart, nervous system, bone, muscle, cartilage, and other organs/tissues.


Sign in / Sign up

Export Citation Format

Share Document