Stem Cell Therapy In Autism: Recent Insights

From Shiapedia

Revision as of 00:01, 28 December 2024 by ZitaEbersbacher (Talk | contribs)
(diff) ← Older revision | Latest revision (diff) | Newer revision → (diff)
Jump to: navigation, search


Correspondence: Dario Siniscalco, Department of Experimental Medicine, University of Campania, Via S. Maria di Costantinopoli 16, 80138 Naples, Italy, Tel +39 eighty one 566 5880, Email dariosin@uab.edu

Collection date 2018.

The full phrases of this license are available at https://www.dovepress.com/phrases.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you hereby settle for the Terms. Non-commercial makes use of of the work are permitted with none additional permission from Dove Medical Press Limited, supplied the work is correctly attributed.

Autism spectrum disorders (ASDs) are characterized by core domains: persistent deficits in social communication and interplay; restricted, repetitive patterns of conduct, interests, or activities. ASDs comprise heterogeneous and complex neurodevelopmental pathologies with nicely-outlined inflammatory situations and immune system dysfunction. As a consequence of neurobiologic adjustments underlying ASD development, cell-primarily based therapies have been proposed and applied to ASDs. Indeed, stem cells show particular immunologic properties, which make them promising candidates in ASD therapy. This comprehensive up-to-date assessment focuses on ASD cellular/molecular abnormalities, potentially helpful stem cell sorts, animal models, and present clinical trials on the usage of stem cells in treating autism. Limitations are also mentioned.

Keywords: autism spectrum disorder, stem cell, cell therapy, immune dysfunction

Autism spectrum disorders (ASDs)

ASDs are very attention-grabbing neurodevelopmental disorders for the medical and scientific group, because of their multifactorial nature and many alternative explanations for their clinical heterogeneity.1 ASD patients show extremely diverse groups of disorders with wide variation in symptoms, intellectual stage, severity, and practical incapacity.2 The variation is due partly to its multifactorial origin that leads ASD to be a neurogenetic clinical entity3,4 with gastrointestinal,5,6 immunologic,7,eight and metabolic implications9 that start within the womb. ASDs are multistage, progressive disorders of mind growth and synapse connections, spanning practically all of pre- and postnatal life.1 ASD starts on the primary embryonic stages with disruption of cell proliferation and differentiation, which ends up in a series of sequential occasions like neural migration, laminar disorganization, altered neuron maturation, neurite outgrowth, issues of synaptogenesis, and lowered neural community functioning.1

ASD impacts greater than 1% of the general population (1:Fifty nine subjects)10 and are characterized by two core signs: the primary one is impaired social communication, and the second situation is restricted, repetitive sorts of behavior, pursuits, or activities. However, the largest problem in autism is triggered by related signs reminiscent of irritability, anxiety, aggression, compulsions, mood lability, gastrointestinal issues, depression, and sleep disorders.11 On the basis of the core and related signs, autism is diagnosed by way of observational and psychometric tests; due to this fact, the clinical prognosis is made primarily based on the presence or absence of core behaviors. The Diagnostic and Statistical Manual of Mental Disorders is conventionally used as a gold normal for autism prognosis.12 However, the neurometabolic variations of autism lead us to search for biologic markers that respond to a right, exact, and concise diagnosis.Thirteen These biologic markers needs to be detected early during pregnancy, as a result of the pathogenesis of ASD just isn't set at one point in time and doesn't reside in a single process, however slightly is a cascade of pre- and postnatal pathogenic processes within the vast majority of ASD toddlers.1

The treatment of ASD is variable and multimodal. It's composed of typical therapies, reminiscent of social abilities training, early intensive conduct therapy, applied habits evaluation, speech therapy, occupational therapy, together with psychotropic drugs,14 transcranial magnetic stimulation,15 and various remedies, which embrace hyperbaric oxygen treatment,16 music therapy, and cognitive and social behavioral therapy.17 Hormonal therapies with oxytocyin have additionally shown some promises in bettering central ASD signs.18 The use of vitamins, herbals, essential oils, and nutritional supplements19,20 and standard therapies have some effect in symptomatic improvement in ASD, although extra research are wanted to confirm these benefits. Developing novel therapies may prove to be the last word intervention for sustained improvement of symptoms in ASD.17 Among the new therapies accessible, there are the gene therapy and stem cell therapy, which have nice potential for treating ASD.21,22 The redesign of mind structure, generated from reprogrammed somatic cells isolated from residing patients, provides new insights into the understanding of autism and thus reverses or ameliorates the symptoms of disorder. Here, we talk about recent advances in using stem cells as a therapy of ASD, in addition to its limitations, implications, and future prospects.

Stem cells for neurologic diseases

The chance to face neurologic diseases and ASD in particular with stem cell utility is described on this part.

Neurologic diseases are often irreversible as a result of sluggish and limited neurogenesis within the mind.23 Therefore, based on the regenerative capability of stem cells, transplantation therapies of various stem cells have been examined in primary research with animal fashions, and preclinical and clinical trials, and plenty of have shown great prospects and therapeutic promises.23 Comparative studies have been raised to grasp nature, properties, and number of donor stem cells, the delivery mode, and the selection of proper affected person populations which will benefit from cell-based therapies.24

However, many occasions these points do not allow to predict why there is no such thing as a appropriate animal mannequin for the research of sure diseases of neurologic growth. Animal fashions of advanced immunogastrometabolic phenomena, such because the ASD, are difficult to validate. The reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) might supply another technique for identifying the cellular mechanisms contributing to autism and the event and testing of many new remedy options.25 This aspect shall be defined at the top of this overview as a result of it comprises the important thing to success of the therapy of stem cells in autism.

Mechanisms underlying therapeutic impact

At the least two key-action mechanisms of stem cells could be helpful for ASD therapy: paracrine effect (the secretome device: cytokines, chemokines, and growth components launched by stem cells and responsible of restore/restoration of injured tissues) and immunomodulatory properties.22 ASDs present immune system abnormalities and strong proinflammatory cytokine production.26,27 ASD topics show an imbalance in innate and adaptive immunity, as CD3+, CD4+, and CD8+ T cells, in addition to pure killer (NK) cells, are altered.27 Immune alterations in ASDs are still confirmed by abnormal monocyte and macrophage responses, which overproduce proinflammatory IL-1β cytokine and show several altered molecular methods resulting in lengthy-time period immune alterations.8,26,28-30 The immune modulatory capability of stem cells may restore these ASD-immune alterations. Stem cells are able to strongly inhibit CD8+ and CD4+ T lymphocytes and NK cell overactivation and proliferation by inhibiting proinflammatory tumor necrosis issue alpha and interferon gamma molecules and rising anti-inflammatory IL-10 levels.31 This fact is confirmed by an animal mannequin of autism, through which stem cells, once transplanted, had been in a position to extend anti-inflammatory cytokine production (detailed in Stem cells and autism: animal models section). The power to modulate the immune system by stem cells is mediated by way of soluble components released by stem cells beneath native chemical setting signaling.32 Another proposed mechanism by which stem cells are in a position to deal with the ASD-mediated immune and inflammatory abnormalities is the cell-to-cell contact activation mechanism, by which stem cells are in a position to switch proinflammatory cells to anti-inflammatory ones.22,32

Table 1 summarizes the harmful processes of ASD that might be contrasted by stem cell’s personal capacities. The next section will explain the potential use of specific stem cell sorts for ASD.

Diagrammatic illustration of different stem cells and associated sources for the remedy of autism and their mechanisms of motion

Note: Homing and mobility capacities are common for all the stem cell types.

Abbreviations: VEGF, vascular endothelial progress issue; HGF, hepatocyte development factor; BDNF, brain-derived neurotrophic issue; NGF, nerve progress factor.

Stem cell sorts

Several kinds of stem cells have been described. The following part shows every sort with potential use for the therapy of ASDs.

Fetal stem cells (FSCs)

FSCs are discovered at numerous phases of human improvement and might be remoted from various somatic organs of fetus, fetal blood, and a wide range of extraembryonic sources like placenta, amniotic membrane, amniotic fluid, and umbilical cord. FSCs have properties that lie in between the embryonic and adult stem cells that they self-renew sooner in culture, have larger differentiation potential and high engraftment price, and don't type teratomas in vivo.33

Fetal SCs from bone marrow categorical pluripotent markers like Oct-4, Nanog, Rex-1, SSEA-3, SSEA-4, Tra-1-60, and Tra-1-81 and show higher telomerase activity, which makes them a preferable candidate for therapeutic applications.33 For treating neurodegenerative diseases, neural stem cells (NSCs) from different components of the fetal mind have been explored. The fetal NSCs have the ability to self-renew and are multipotent with the ability to differentiate into neurons, astrocytes, and oligodendrocytes.34-36 FSCs have the flexibility to secrete various neurotrophic and immunomodulatory components that promote neuronal progress and suppress the motion of proinflammatory cytokines that make them a potential candidate for treating ASDs and various neurodegenerative diseases.37

There are various experiences on the successful use of FSCs for different neurodegenerative disorders. When the fetal NSCs were transplanted into the rat fashions of temporal lobe epilepsy and ischemia, they get grafted and differentiated into neurons.34,35 A clinical trial in youthful human Parkinson’s patients additionally confirmed promising outcomes when dopamine neurons from fetal neural tissues have been transplanted.36 An open-labeled clinical trial that examined the security and efficacy of FSCs in autistic kids revealed no antagonistic occasions and a major distinction in the development of autistic symptoms (detailed in Stem cells and autism: animal fashions section).37

Bone marrow-derived stem cells

Mesenchymal stem cells (MSCs)

MSCs are present in nearly all of the adult tissues and are characterized by their ability to adhere to plastic surface, express cell floor antigens like CD105, CD90, and CD73, not express CD34, CD45, CD14 or CD11b, CD79α or CD19, and HLA-DR, and differentiate into osteoblasts, chondroblasts, and adipocytes in tradition.38 Aside from these minimal characteristics, MSCs have wider properties like their skill for rapid proliferation, differentiation into cell forms of endodermal and ectodermal origins, secretions of various tropic components, and immunomodulatory action, which make them a preferable candidate for cellular therapies.39,40

The paracrine secretion and immunomodulatory properties of MSCs make them a possible candidate for treating ASDs.Forty one Cultured MSCs secrete various neurotrophic elements including vascular endothelial development factor (VEGF), hepatocyte progress issue, mind-derived neurotrophic issue (BDNF), and nerve progress issue and exhibit neuroprotective effects.Forty two MSCs are additionally hypoimmunogenic and have immunosuppressive properties that they don't express co-stimulatory molecules corresponding to CD80, CD86, or CD40 and specific low levels of MHC class I molecules.43,44 Different mechanisms have been hypothesized for using MSCs for treating ASDs that embody inducing plasticity, secretion of anti-inflammatory and survival-selling components, and engrafting into neural community.Forty five

Also, in vivo, in numerous animal fashions of neurodegeneration, MSCs exert neuroprotection mainly by secretion of varied neurotrophic and immunomodulatory factors, thus facilitating the recruitment of endogenous stem cells to promote regeneration and by downregulating T cells, B cells, and NK cells of immune system.Forty six Owing to these properties, MSCs have been extremely most well-liked candidates for clinical trials for varied neurologic diseases. Clinical trials utilizing MSCs are ongoing for diseases like a number of sclerosis (MS), stroke, Parkinson’s illness (PD), Huntington’s disease (HD), Alzheimer’s illness (Ad), and systemic autoimmune diseases.Forty seven For ASDs, several research utilizing stem cells have been carried out in people (detailed in Stem cells and autism: animal models part); a research by Lv et al analyzed the safety and efficacy of utilizing human cord blood mononuclear cells (CBMNCs) and umbilical cord-derived mesenchymal stem cells (UCMSCs) in treating youngsters with autism.Forty eight They reported safe and statistically vital enhancements in the Childhood Autism Rating Scale (Cars), Clinical Global Impression (CGI) scale, and Aberrant Behavior Checklist (ABC) within the children handled combinedly with CBMNCs and UCMSCs when compared to the control group.

Adipo-stem cells

Stem cells can also be isolated from completely different adipose tissues within the body in a minimally invasive method and are termed as adipose-derived stem cells (ASCs). Just like MSCs, ASCs are plastic adherent, express CD90, CD105, CD73, CD44, and CD166, lack the expression of CD45 and CD34, and have the flexibility to differentiate into cells of all three germ layers.49,50

Together with being multipotent, ASCs secrete various trophic components and are immunosuppressive and hypoimmunogenic, making them a sexy candidate for cellular therapies. ASCs have been utilized in numerous clinical trials focusing on a variety of indications starting from immune disorders, myocardial infarction, bone defects, and neurodegenerative diseases. Although no reports have been found on using ASCs for treating ASDs, ASCs have been confirmed efficient in different neurologic preclinical fashions, as within the mouse mannequin of center cerebral artery occlusion, human ASCs partially rescue the stroke syndromes by forming new neurons and blood vessels and rising the viability of endogenous neurons.Fifty one

Umbilical cord- and amniotic fluid-derived stem cells

Umbilical cord and placenta also provide a great source of stem cells.22 These stem cells offer robust potential therapeutical applications and show mesenchymal traits.Fifty two The Wharton Jelly of the umbilical cord and the amniotic fluid are other sources of stem cells possessing high in vitro progress capacity and low immunogenicity and immunomodulation properties, expressing the standard antigen profile of MSCs.53,54 These cells from perinatal, extraembryonic tissue have potential for future applications in ASDs.22 There are not any moral controversy and danger of teratoma formation, and they may be used for autologous transplantation after banking in later levels of life.Fifty five

NSCs

NSCs are discovered within the fetal and grownup human brain and have the potential for extensive proliferation and differentiation into three major cell varieties of the nervous system, the neurons, astrocytes, and oligodentrocytes.56 The NSCs that may be extracted from two major regions of the brain, specifically, the subventricular zone of lateral ventricles and subgranular zone of hippocampus, will be cultured.57,58 The culture-expanded NSCs are multipotent, have the flexibility to differentiate into varied neuronal cell sorts, secrete neurotrophic elements, combine into neural tissue, maintain homeostasis, and are neuroprotective, making them a perfect candidate for treating ASDs. Indeed, impairments in excitatory and inhibitory cortical neurons lead to minicolumn structure abnormalities in ASDs.Fifty six Synaptic-associated genes show a number of uncommon variants in some ASD subjects.59 Given that, transplantation of NSCs might be efficient in ASDs, as transplanted cells can promote neural tissue repair and homeostasis through integration in broken areas and secretion of factors that improve brain restore and plasticity.60 The definitive use of NSCs for clinical functions in neurodegenerative diseases still requires addressing some vital points: autologous dependable supply of adequate quantity of stem cells must be recognized; put up-transplanted neural plasticity and differentiation, if any, must be further outlined.60 However, though NSCs have been used in various preclinical and clinical research in opposition to totally different neurologic conditions like PD, HD, Ad, amyotrophic lateral sclerosis (ALS), MS, stroke, and spinal cord damage (SCI), the end result just isn't definitive as anticipated and is hindered by a number of points to be additional elucidated: the absence of homogenous cell inhabitants, stability, and long-term survival of neurons after transplantation.61,sixty two

Different strategies have been tried to reinforce the capabilities of NSCs, like immortalizing the NSCs by gene manipulation techniques to create a homogenous, lengthy-surviving cell that is capable of differentiating into neurons and glial cells when transplanted into regular or damaged mind.Fifty eight

Hematopoietic stem cells (HSCs)

HSCs are primarily resident in bone marrow and in addition in blood and umbilical cord. Specific cluster of differentiation markers characterize them (CD34, CD59, CD117, CD133, CD164).Sixty three Self-renewal, multipotency, and homing/mobility activities are very high. This kind of stem cells is able to differentiate in myeloid and lymphoid lineages. Their paracrine activity, releasing bioactive molecules, and their skill to quickly site visitors to the location of inflammation gained them lots of attention for his or her use in ASD therapy.Sixty three Several clinical trials have been carried out with using CD34+ stem cells in autism (detailed in Stem cells and autism: animal fashions part).

iPSCs: the new frontier for cell therapy

iPSCs may represent a novel instrument for ASD therapy.

iPSCs are comparatively new kind of stem cells that are reprogrammed from any cell kind of the physique by ectopic expression of various transcriptional elements initially by using integrating vectors like retroviruses and lentiviruses. The iPSCs derived by these strategies pose threat of insertional mutagenesis and make them unsafe for therapeutic applications. To beat this impediment, various new methods like use of small molecules, sendai virus, adenovirus, RNA molecules synthesized by plasmids, and recombinant proteins have developed to create integration-free iPSCs.64 The iPSCs derived from different strategies have the traits similar to that of embryonic stem cells, with the ability to self-renew, specific stem cell markers, and differentiate into the cell of all the three germ layers except the cells in further-embryonic tissue. The principle benefit of iPSCs over the ES cells is their easy means of derivation from any cell type without the need for embryos and that they serve as a limiteless cell supply for autologous therapy. iPSCs derived from the donors have many applications other than being used for regenerative therapies, and the patient-specific iPSCs are used in disease modeling, toxicity testing, and drug screening research.64

Disease-particular iPSCs derived from patients function an infinite source for drug testing research. A wide range of iPSCs are produced from patients suffering from unifactorial and multifactorial diseases starting from adenosine deaminase deficiency-associated severe combined immunodeficiency, PD, HD, ALS, Down syndrome, kind 1 diabetes, Duchenne muscular dystrophy, spinal muscular atrophy, and β-thalassemia65 that serve as a model for disease and for drug testing on them.

With regard to therapeutic utilization, iPSCs are seen as a possible candidate for treating numerous disorders. The proof to substantiate the usage of iPSCs for clinics comes from varied in vitro and in vivo preclinical trials. In in vitro, iPSCs can be made to differentiate into various cell varieties like dopamine and motor neurons, dendritic cells, practical cardiomyocytes, macrophages, hepatocytes, and hematopoietic and endothelial cells.Sixty six There are a number of in vivo research that show the efficiency of iPSCs in treating neurodegenerative disorders, cardiovascular disease, and sickle cell anemia. For example, in contusive SCI model in nonobese diabetic extreme combined immunodeficient mice, injection of neurospheres derived from human iPSCs leads to recovery of locomotor perform with out formation of any tumors.67 The differentiation capabilities of injected neurospheres into neurons, astrocytes, and oligodendrocytes together with induction of angiogenesis, axonal regeneration, and native-circuitry reconstruction might contribute to the recovery in SCI model.67 When autologous iPSCs-derived dopamine neurons have been transplanted in PD model of cynomolgus monkey, the neurons engraft and survive for prolonged time of two years leading to improvement in the motor operate in the nonhuman primate model.Sixty eight iPSCs have additionally been used effectively against HD and ALS by their capability to differentiate into desired neuronal lineages.69,70

Despite the promising in vitro and preclinical studies, the usage of iPSCs in clinical trials continues to be in infant stage. There are some major challenges like tumorigenicity and immunogenicity to be addressed before utilizing iPSCs for clinical functions. iPSCs used for transplantation may lead to teratoma formation: any residual pluripotent stem cells in grafting experiment might induce teratoma. This issue might be addressed by developing more efficient nonintegrative approaches for deriving iPSCs, using differentiated cells somewhat than iPSCs and likewise by prior screening and collection of cells before transplantation.71 For immunogenicity concern, though mostly iPSCs are used in autologous fashion there are some evidences that showed that iPSCs and cells derived from iPSCs can nonetheless elicit little immune response in syngeneic recipients that again could be overcome by improved reprogramming applied sciences.Seventy two

iPSC purposes acquire much consideration additionally for autism analysis.Seventy three Successful reprogramming of peripheral blood-derived mononuclear cells from autistic baby into iPSCs has been performed by transgene-free supply system.74 Customized iPSCs will assist in elucidating the pathogenic mechanisms of ASDs,seventy five also for neuronal differentiation and maturation.76 Indeed, iPSC-derived neurons from autistic topics present aberrant cation channels expression, voltage-gated currents, and changes in synaptic functions.77,78 Autistic patient-derived stem cells display an altered developmental neuronal phenotype: alteration in cell bodies, branched neurites, and motility in contrast with these derived from controls.Seventy nine Using iPSCs to generate three-dimensional fashions of neurons and mind constructions may be helpful to mannequin autism pathophysiology.80

Stem cells and autism: animal fashions

Animal fashions are playing a crucial position in exploring the potential of stem cells for growing therapeutic strategies for ASDs and will provide novel insights by way of which stem cells can help in ameliorating ASD-associated phenotypes. These data cannot be achieved by clinical trials.

As lifelong neurodevelopmental pathologies, ASDs are uniquely human behaviors and animal fashions fail to totally reproduce the human situation; however, they might present some essential primary data.81 A commonly used rodent model for ASD is the black and tan brachyuric (BTBR) inbred mouse strain. BTBR mice spontaneously develop behavioral deficits and brain abnormalities reflecting ASDs. Human MSCs have been intracerebroventricularly transplanted (50,000 cells/µL) in BTBR mice.Eighty two Cell therapy improved repetitive behaviors in transplanted mice by way of decreasing digging and self-grooming duration and growing the latency between two consecutive occasions. Cell-transplanted mice additionally displayed lowering in cognitive rigidity as measured by water T-maze take a look at. To note, social habits was additionally improved: transplanted BTBR mice reported higher social approach and social novelty preference with respect to nontransplanted BTBR mice.Eighty two By a cellular point of view, hMSCs improved hippocampal neurogenesis (growing of Ki-67 and DCX markers) and increased BDNF levels; after 6 weeks put up-transplantation, hMSCs were found to be located close to the wall of the dorsal third ventricle. In the identical mouse model, hMSC transplantation (50,000 cells/µL into the cerebral lateral ventricle) confirmed lengthy-time period beneficial results in ameliorating autistic-like symptoms.83 Social behaviors were improved and stereotypic behaviors have been reduced at 6 months posttransplantation. Another human stem cell type, the ASCs (50,000 cells/µL, intraventricularly), was efficiently utilized in valproic acid (VPA)-induced autism mouse mannequin.Eighty four Transplanted mice displayed increased motor coordination (as measured by open-discipline take a look at) and social behaviors and decreased anxiety. Human ASCs were accountable to extend the phosphatase and tensin homolog, VEGF, IL-10 expression, and p-AKT/AKT ratio in the brains of VPA mice.Eighty four Mouse-derived MSCs have been utilized in VPA-induced autism mannequin, leading to increasing neurogenesis and promoting maturation of newly formed neurons within the dentate gyrus 2 months after transplantation.85 MSCs were expanded in tradition for up to 20 passages and immunophenotyped for mesenchymal particular markers. Then hundreds of MSCs in 5 µL MSC were transplanted into the precise lateral ventricle. Deficits in cognitive and social behaviors were additionally improved after 2 weeks posttransplantation.85

Beyond stem cell therapy as priceless software for treating autism, stem cells additionally supply the possibility for in-depth study of ASD pathology. Offspring of immune-activated mothers (maternal immune activation autism model) develop preferential myeloid lineage potential and altered differentiation of HSCs.86 The importance of this work displays that immune changes throughout maternal life could confer alterations in stem cells lineage by means of your complete life of offspring.

Transcriptome evaluation revealed differential expressed genes in dental pulp stem cells from idiopathic ASD subjects compared to controls.87 While stem cells could possibly be used for understanding the biologic mechanisms of ASDs, the cellular changes of ASD-derived stem cells pose some concerns for autologous cell transplantation, as described additional on this overview.

Stem cells and autism: clinical trials

Clinical trials on stem cell transplantation in ASDs are of important significance to validate safety (in first) and the efficacy of the cellular therapy.

Currently, a number of clinical trials have been performed to display security and efficacy of stem cells autism management (Table 2). Sharma et al conducted an open-label proof-of-concept examine on the use of autologous bone marrow-derived mononuclear cell (BMMNC) transplantation in 32 topics (median age at intervention 10.5 years, male:feminine 3:1) with confirmed diagnosis of autism.88 The protocol included intrathecal cellular therapy followed by occupational therapy, sensory integrative method, speech therapy, psychological intervention, and specific dietary suggestions. Mononuclear cell fraction is a heterogeneous mixture consisting of endothelial progenitors, HSCs and MSCs, and multipotent grownup progenitor cells. These cells were separated from the aspirate of bone marrow, counted by CD34+ marker and checked for viability. On the same day, the cells were intrathecally injected and methyl prednisolone was given intravenously to enhance survival of the injected cells. Long-time period hostile events have been monitored to establish the safety of stem cell transplantation. Minor opposed events (vomiting, nausea, ache at aspect of injection, or aspiration) were present. These events had been reported to be procedure-associated and not cellular transplantation-associated issues. Minimal improve in hyperactivity was recorded as a serious hostile event related to cell procedure, together with three patients growing seizures. After cellular therapy, Indian Scale for Assessment of Autism (ISAA) indicated improvements in the domains of social relationships and reciprocity (improved eye contact, social smile, and reaching out to others), cognitive points (attention, focus, and time of response), and in addition in speech and language patterns (reduction in echolalic speech, engaging in stereotyped repetitive use of language, manufacturing of infantile squeals or unusual noises, inability to initiate or sustain dialog with others, inability to understand the pragmatics of the conversation, and speech regression). Decrease in inappropriate emotional responses, exaggerated emotions, engaging in self-stimulating feelings, and getting excited or agitated for no obvious cause was recorded. CGI scale scored changes within the severity of the illness, overall enchancment, and the efficacy of the therapy. Interestingly, useful neuroimaging, by the technique of positron emission tomography (PET) scan, showed elevated 18F-fluorodeoxyglucose (18F-FDG) uptake within the areas of frontal lobe, cerebellum, amygdala, hippocampus, parahippocampus, and mesial temporal lobe after 6 months of cellular therapy. Hypoperfusion has been just lately demonstrated in key mind areas of ASD subjects.89 Hypometabolic areas before cell transplantation showed increased metabolism after cellular supply, probably resulting from improved oxygenation and functioning of the broken neurons. As limitations, authors report the small pattern measurement, and the absence of randomization and a control group.88

Table 2.

Clinical trials carried out on the use of stem cells in ASDs

Most recently, Sharma et al performed the sort of cellular remedy on a male autistic grownup (25 years old).90 Autologous BMMNCs were intrathecally transplanted. No major opposed occasions had been seen. After 6 months of cellular therapy, enhancements in concentration, sitting tolerance, consideration, sleep, eye contact, social interactions, and reminiscence have been reported, as measured by ISAA, Cars, and Functional Independence Measure scores before and after cellular therapy. Six months posttherapy, 18F-FDG PET scan showed enhancements in mind hypometabolism.

Lv et al carried out a Phase I/II trial to research the security and efficacy of combined transplantation of CBMNCs and UCMSCs in treating children with autism.Forty eight Thirty-seven autistic youngsters (just one female) were recruited and randomly subdivided in control (imply age 5.60) and experimental teams. This group was further divided into CBMNC group (imply age 7.4) and combined group (mean age 6.2) which obtained each forms of cells. No adverse effects were recorded on the time of transplantation and during the entire follow-up. As minor results, only five kids developed a transiently low-grade fever. About cell process, the cells had been remoted from gifted cord blood and umbilical cord of knowledgeable healthy donors. CD34+ CBMNCs were extracted and in vitro grown in a GMP facility. After extraction from umbilical cord, UCMSCs were in vitro expanded and labeled for MSC characterization: CD29+, CD73+, CD90+, CD105+, the expression of CD45, CD34, CD14, CD79, and HLA-DR was absent or
In one other open-label pilot examine, a total of 45 autistic youngsters (39 males, six females, mean age 6.9) had been enrolled to evaluate the safety of the use of FSCs in ASDs.37 FSCs were harvested from 5- to 9-week-old human fetuses (one donor pattern for every transplantation procedure) following voluntarily selective pregnancy terminations. Donor women and fetal samples had been tested for bacterial, fungal, and viral infections and parasites. CD34+ HSCs have been extracted from fetal liver (a robust niche for HSCs)91 and mind and in vitro expanded. Colony-forming items and neurosphere formation had been checked. HSCs were then saved in liquid nitrogen at −196°C within the cryobank and were intravenously transplanted at day 1. On the second day, the fetal cells were injected into the subcutaneous abdominal adipose tissues. ASD kids handled with FSCs displayed no adverse occasions or unwanted side effects due to cell therapy or steroid and antihistamine combination pretreatment through the 1-yr comply with-up interval studied. The early submit-cellular transplantation results observed have been improved eye contact, appetite, and socialization. Autism Treatment Evaluation Checklist and ABC scores at follow-up instances of 6 and 12 months confirmed improvements in sociability, cognitive capability, and behaviors, particularly at the ultimate time level. To notice, pre- and put up-cellular remedy immunologic findings indicated an improved cell-mediated immunity, resulting from a major improve of CD3+ T lymphocytes and CD4+ T helpers, and a lower in CD19+ B lymphocytes counts after the cellular therapy (pretreatment values had been abnormal). Fetal SC transplantation was protected and nicely tolerated. Based on the authors, the administration routes (intravenous and subcutaneous) have been chosen on the premise of their much less invasivity. Immunomodulatory features of FSCs seem to be a key mechanism of action.37

Recently, Phase I/II results of a clinical trial on autologous umbilical cord blood (AUCB) transplantation in idiopathic ASD youngsters had been published.Ninety two As crossover examine, ASD youngsters (age 2-7 years), with umbilical cord blood cryopreserved and saved, had been transplanted with both AUCB or placebo (0.9% saline solution), and evaluated at baseline, 12, and 24 weeks. The topics have been randomly divided into two groups. The first group acquired cord blood infusion and then saline injection. The second group acquired saline infusion first, then cord blood infusion. Accordingly to the authors, the selection of the crossover study is that in this manner each topic serves as its personal management. As well as, the crossover design resulted in wonderful compliance by dad and mom. Transplanted ASD youngsters were evaluated at 12 and 24 weeks posttransplantation. UCB was checked for sterility, viability, colony-forming unit, and CD34+ cell rely. Some doable UCB therapy-associated gastrointestinal disorders were reported. No severe antagonistic occasions required treatment. CGI and Expressive/Receptive One Word Picture Vocabulary Tests scores reported minimal proof of clinical effectiveness of AUCB therapy. While crossover research offers the possibility to minimize the baseline variability, as limitations put up-thaw CFU capacity and CD34+ count must be better addressed. Another point of discussion is the precise AUCB dosage, various amongst enrolled subjects because of parental alternative in using own biobanked UCB. This latter study reveals conflicting outcomes in comparison with the Dawson examine.93 Twenty-5 ASD children (21 males, 4 females, mean age 4.6 years) received a single intravenous infusion of AUCB. Sterility, infection illness markers, nucleated cell depend, CD34+ depend, and CFU had been tested on biobanked UCB. A portion of or your complete own cord blood unit (1-5 × 107 cells/kg) was infused per ASD topic. AUCB infusion was properly tolerated, as no serious adversarial effect was reported in any participant. Agitation, allergy, and pores and skin adjustments have been reported as unrelated hostile results. Vineland Adaptive Behavior Scale-II, CGI, Pervasive Developmental Disorder Behavior Inventory, and Expressive One-Word Picture Vocabulary Test-4 had been the main assessments used to assess cognitive and behavioral enhancements. Socialization, communication, and adaptive habits scores were improved at 6 months postinfusion and had been sustained between 6 and 12 months posttreatment.Ninety three Interestingly, nonverbal, greater IQ ASD children confirmed larger enhancements in behaviors. However, as authors said, the observed behavioral outcomes may very well be as a result of natural course of improvement during the preschool period.

Limitations

Several limitations and recommendations have to be taken under consideration before claiming definitive outcomes about cellular therapy in ASD.

Stem cell therapy offers novel alternative to develop cell-based drugs and purposes to treat several diseases, including immune and neurodegenerative pathologies,ninety four in addition to ASD.22 It is essential that cellular therapy is carried out underneath laboratory and clinical guidelines. The International Association of Neurorestoratology has recently revealed the Clinical Cell Therapy Guidelines for Neurorestoration.Ninety five These tips concern the recommended requirements for personnel, amenities, and establishments that carry out cell-based mostly therapies, as well as the moral requirements, and include the provisions: affected person-informed consent/IRB; indications/contraindications for cell therapy; documentation of process and therapy; security and efficacy evaluations; coverage of repeated therapies; not charging patients for unproven therapies; basic principles of cell therapy; and publishing responsibility.Ninety five While the mechanisms of action of stem cells are being defined by primary research, in addition to by in vivo models, clinical trials present scientific effectiveness for human purposes.96 However, from the clinical trials examined above (Table 2), a number of limitations have to be taken into account, earlier than claiming definitive outcomes for using stem cells in ASD treatment.

Relatively small number of topics

The variety of enrolled subjects requires bigger studies on using the cell therapy in ASDs. All the analyzed clinical trials admit the small pattern size used. Control group additionally needs to be more standardized.

Time of comply with-up

Harmonization of the time of the research is desirable. Long-time period outcomes or the selection of multiple well timed transplantations have to be elucidated.

Efficacy

The efficacy of cell therapies in ASDs ought to be evaluated by internationally validated, standardized, and ideally harmonized scales. Too many various scales and scores, even when properly adopted and established by international scientific community, usually do not show the efficient result of cell transplantation.

Cell heterogeneity and culturing

A greater characterization of cell sorts utilized in all the abovementioned research will assist in the right choice of cell to be utilized. If the cells require in vitro growth earlier than transplantation remains to be clarified. To notice, two clinical studies92,93 used part of or whole AUCB. Eventually cellular senescence and life span need extra clarification. Senescence of cultured stem cells is another main concern for clinical trials. Prolonged in vitro expansion could trigger senescence phenotype and telomere shortening, in this way the stem cell regenerative capacity could be modified.Fifty four

Cell source and dosage

As seen in the research summarized in Table 2, the cell sources are very totally different, with significant properties varying from tissue to tissue. Proliferation and enlargement potential are affected additionally by donor age.Ninety seven The efficient cell dosage must be decided extra. The optimal stem cell sort for cellular grafts is still uncertain.

Autologous or allogenic transplantation?

The choice between autologous or allogenic transplantation remains a key issue.98 Autologous infusion requires preventive storage of the personal biomaterial, with the associated issues (ie, the fee for the cryostorage). As well as, even when some inherited genetic diseases are treated with stem cells (ie, thalassemia main or severe mixed immune deficiency [SCID] diseases),ninety nine the likelihood to treat the circumstances of ASDs attributable to genetic defects (nonsyndromic autism)100 is still debated. If some genetic modifications are current in the ASD topic, these modifications can be saved by all the saved and biobanked cell varieties. Allogenic transplantation overcomes this latter problem. However, the availability of donors and subsequent infectious disease assessments are key points to be taken under consideration.

Route of cell administration

Intrathecal route seems to be invasive. However, authors who used this way of administration reported its security and efficient delivery of cells to mind, as transplanted cells could reach vital central nervous system areas by means of cerebrospinal fluid, enhancing cell homing onto damaged centers.A hundred and one

Ethics

Ethics in stem cell treatments, as well as in ASD administration, require particular attention.102 Cell therapy have to be performed below regulation and approval of regulatory establishments. Within the case of unfavorable results from clinical trials, follow of the cell therapy ought to be discontinued.

Another question is the validity of repeated cell remedies. Presently, there is no such thing as a published proof of the effectiveness of repeated cell transplantations in ASDs and clinical trials needs to be performed. Dawson et al spotlight that the financial status could have an effect on cell therapy availability.93 Especially in the case of AUCB, some ethnic teams might not be able to deal the price of biobanking.

Preclinical evidences spotlight potential profit and necessary advances for the usage of cellular therapy in ASDs. As the mechanisms by which transplantation of stem cells results in an enhanced functional restoration and structural reorganization must be better elucidated,61 in vivo research must present definitive outcomes on the mechanism of action of stem cells that in some circumstances it isn't potential to reproduce or research in humans (ie, detailed analysis of secretome process). Initially, the first concept on mechanism of action of stem cells was that cell therapy may act by a "cell replacement" mechanism; nowadays, large emerging evidences have proven that cell therapy works by offering trophic or "chaperone" help to the injured tissue and mind.103 It is noteworthy to think about that the clinical trials on stem cell transplantation in ASD topics clearly point out the security of the procedures (safety is the primary final result). These research additionally report important encouraging optimistic effects in relief of ASD symptoms (part of the work of Chez et al,92 as talked about earlier, where the authors claim just for a trend of amelioration) and advocate for the usage of cellular therapy in ASDs. However, to date, only five clinical trials have been carried out (Table 2) with a number of differences among them (study design, subjects enrolled, cellular sorts, route of administration, end result measures), which will require further examinations. Taken collectively the limitation concerns and the promising ameliorative effects of cellular therapies in ASD therapy, extra full and exhaustive investigations and huge trials will probably be wanted so as to assert definitive outcomes.104

Footnotes

Disclosure

The authors report no conflicts of interest on this work.

- 1.Courchesne E, Pramparo T, Gazestani VH, Lombardo MV, Pierce K, Lewis NE. The ASD residing biology: from cell proliferation to clinical phenotype. Mol Psychiatry. 2018June:22. doi: 10.1038/s41380-018-0056-y. Epub. [DOI] [PMC free article] [PubMed] [Google Scholar]- 2.Geschwind DH. Advances in autism. Annu Rev Med. 2009;60:367-380. doi: 10.1146/annurev.med.60.053107.121225. [DOI] [PMC free article] [PubMed] [Google Scholar]- 3.Chaste P, Leboyer M. Autism risk elements: genes, environment, and gene-surroundings interactions. Dialogues Clin Neurosci. 2012;14(3):281-292. doi: 10.31887/DCNS.2012.14.3/pchaste. [DOI] [PMC free article] [PubMed] [Google Scholar]- 4.Mottron L, Belleville S, Rouleau GA, Collignon O. Linking neocortical, cognitive, and genetic variability in autism with alterations of mind plasticity: the Trigger-Threshold-Target model. Neurosci Biobehav Rev. 2014;47:735-752. doi: 10.1016/j.neubiorev.2014.07.012. [DOI] [PubMed] [Google Scholar]- 5.Iovene MR, Bombace F, Maresca R, et al. Intestinal dysbiosis and yeast isolation in stool of topics with autism spectrum disorders. Mycopathologia. 2017;182(3-4):349-363. doi: 10.1007/s11046-016-0068-6. [DOI] [PubMed] [Google Scholar]- 6.Rose DR, Yang H, Serena G, et al. Differential immune responses and microbiota profiles in youngsters with autism spectrum disorders and co-morbid gastrointestinal symptoms. Brain Behav Immun. 2018;70:354-368. doi: 10.1016/j.bbi.2018.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]- 7.Onore C, Careaga M, Ashwood P. The role of immune dysfunction in the pathophysiology of autism. Brain Behav Immun. 2012;26(3):383-392. doi: 10.1016/j.bbi.2011.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]- 8.Siniscalco D, Schultz S, Brigida AL, Antonucci N. Inflammation and neuroimmune dysregulations in autism spectrum disorders. Pharmaceuticals (Basel) 2018;11(2):E56. doi: 10.3390/ph11020056. [DOI] [PMC free article] [PubMed] [Google Scholar]- 9.Ghaziuddin M, Al-Owain M. Autism spectrum disorders and inborn errors of metabolism: an update. Pediatr Neurol. 2013;49(4):232-236. doi: 10.1016/j.pediatrneurol.2013.05.013. [DOI] [PubMed] [Google Scholar]- 10.Baio J, Wiggins L, Christensen DL, et al. Prevalence of autism spectrum disorder among youngsters aged 8 years - autism and developmental disabilities monitoring network, 11 sites, united states, 2014. MMWR. Surveill Summ. 2018;67(6):1-23. doi: 10.15585/mmwr.ss6706a1. [DOI] [PMC free article] [PubMed] [Google Scholar]- 11.Elsabbagh M, Divan G, Koh YJ, et al. Global prevalence of autism and different pervasive developmental disorders. Autism Res. 2012;5(3):160-179. doi: 10.1002/aur.239. [DOI] [PMC free article] [PubMed] [Google Scholar]- 12.American Psychiatric Association . Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Washington DC: American Psychiatric Association; 2013. [Google Scholar]- 13.Pratt J, Hall J. Biomarkers in neuropsychiatry: a prospect for the twenty-first Century? Curr Top Behav Neurosci. 2018 Jul 3;7:1-8. doi: 10.1007/7854_2018_58. Epub. [DOI] [PubMed] [Google Scholar]- 14.Thibaut F. New perspectives in autism spectrum disorders. Dialogues Clin Neurosci. 2017;19(4):323. doi: 10.31887/DCNS.2017.19.4/fthibaut. [DOI] [PMC free article] [PubMed] [Google Scholar]- 15.Oberman LM, Rotenberg A, Pascual-Leone A. Use of transcranial magnetic stimulation in autism spectrum disorders. J Autism Dev Disord. 2015;45(2):524-536. doi: 10.1007/s10803-013-1960-2. [DOI] [PMC free article] [PubMed] [Google Scholar]- 16.Rossignol DA, Bradstreet JJ, Van Dyke K, et al. Hyperbaric oxygen remedy in autism spectrum disorders. Med Gas Res. 2012;2(1):16. doi: 10.1186/2045-9912-2-16. [DOI] [PMC free article] [PubMed] [Google Scholar]- 17.Sharma SR, Gonda X, Tarazi FI. Autism spectrum disorder: classification, diagnosis and therapy. Pharmacol Ther. 2018;190:91-104. doi: 10.1016/j.pharmthera.2018.05.007. [DOI] [PubMed] [Google Scholar]- 18.DemMayo MM, Song YJC, Hickie IB, Guastella AJ. A assessment of the safety, efficacy and mechanisms of supply of nasal oxytocin in youngsters: therapeutic potential for autism and Prader-Willi syndrome, and proposals for future research. Paediatr Drugs. 2017;19(5):391-410. doi: 10.1007/s40272-017-0248-y. [DOI] [PubMed] [Google Scholar]- 19.Gasparotto FM, Dos Reis Lívero FA, Tolouei Menegati SEL, Junior AG. Herbal medicine in its place treatment in autism spectrum disorder: a systematic evaluate. Curr Drug Metab. 2018;19(5):454-459. doi: 10.2174/1389200219666171227202332. [DOI] [PubMed] [Google Scholar]- 20.Adams JB, Audhya T, Geis E, et al. Comprehensive nutritional and dietary intervention for autism spectrum disorder-a randomized, controlled 12-month trial. Nutrients. 2018;10(3):E369. doi: 10.3390/nu10030369. [DOI] [PMC free article] [PubMed] [Google Scholar]- 21.Benger M, Kinali M, Mazarakis ND. Autism spectrum disorder: prospects for remedy utilizing gene therapy. Mol Autism. 2018;9:39. doi: 10.1186/s13229-018-0222-8. [DOI] [PMC free article] [PubMed] [Google Scholar]- 22.Siniscalco D, Bradstreet JJ, Sych N, Antonucci N. Perspectives on using stem cells for autism therapy. Stem Cells Int. 2013;2013:262438. doi: 10.1155/2013/262438. [DOI] [PMC free article] [PubMed] [Google Scholar]- 23.Song CG, Zhang YZ, Wu HN, et al. Stem cells: a promising candidate to deal with neurological disorders. Neural Regen Res. 2018;13(7):1294-1304. doi: 10.4103/1673-5374.235085. [DOI] [PMC free article] [PubMed] [Google Scholar]- 24.Mazzini L, Ferrari D, Andjus PR, et al. BIONECA Cost Action WG Neurology Advances in stem cell therapy for amyotrophic lateral sclerosis. Expert Opin Biol Ther. 2018;18(8):865-881. doi: 10.1080/14712598.2018.1503248. [DOI] [PubMed] [Google Scholar]- 25.Brick DJ, Nethercott HE, Montesano S, et al. The autism spectrum disorders stem cell resource at Children’s Hospital of Orange County: implications for illness modeling and drug discovery. Stem Cells Transl Med. 2014;3(11):1275-1286. doi: 10.5966/sctm.2014-0073. [DOI] [PMC free article] [PubMed] [Google Scholar]- 26.Siniscalco D, Mijatovic T, Bosmans E, et al. Decreased numbers of CD57+CD3- cells determine potential innate immune variations in patients with autism spectrum disorder. In Vivo. 2016;30(2):83-89. [PubMed] [Google Scholar]- 27.Inga Jácome MC, Morales Chacòn LM, Vera Cuesta H, et al. Peripheral inflammatory markers contributing to comorbidities in autism. Behav Sci (Basel) 2016;6(4):E29. doi: 10.3390/bs6040029. [DOI] [PMC free article] [PubMed] [Google Scholar]- 28.Brigida AL, Schultz S, Cascone M, Antonucci N, Siniscalco D. Endocannabinod signal dysregulation in autism spectrum disorders: a correlation link between inflammatory state and neuroimmune alterations. Int J Mol Sci. 2017;18(7):E1425. doi: 10.3390/ijms18071425. [DOI] [PMC free article] [PubMed] [Google Scholar]- 29.Siniscalco D, Sapone A, Giordano C, et al. The expression of caspases is enhanced in peripheral blood mononuclear cells of autism spectrum disorder patients. J Autism Dev Disord. 2012;42(7):1403-1410. doi: 10.1007/s10803-011-1373-z. [DOI] [PubMed] [Google Scholar]- 30.Siniscalco D, Sapone A, Giordano C, et al. Cannabinoid receptor kind 2, however not type 1, is up-regulated in peripheral blood mononuclear cells of children affected by autistic disorders. J Autism Dev Disord. 2013;43(11):2686-2695. doi: 10.1007/s10803-013-1824-9. [DOI] [PubMed] [Google Scholar]- 31.Caprnda M, Kubatka P, Gazdikova K, et al. Immunomodulatory effects of stem cells: therapeutic choice for neurodegenerative disorders. Biomed Pharmacother. 2017;91:60-69. doi: 10.1016/j.biopha.2017.04.034. [DOI] [PubMed] [Google Scholar]- 32.Siniscalco D, Giordano C, Galderisi U, et al. Long-lasting effects of human mesenchymal stem cell systemic administration on pain-like behaviors, cellular, and biomolecular modifications in neuropathic mice. Front Integr Neurosci. 2011;5:79. doi: 10.3389/fnint.2011.00079. [DOI] [PMC free article] [PubMed] [Google Scholar]- 33.Guillot PV, Gotherstrom C, Chan J, Kurata H, Fisk NM. Human first-trimester fetal MSC specific pluripotency markers and develop sooner and have longer telomeres than adult MSC. Stem Cells. 2007;25(3):646-654. doi: 10.1634/stemcells.2006-0208. [DOI] [PubMed] [Google Scholar]- 34.Lee H, Yun S, Kim IS, et al. Human fetal mind-derived neural stem/progenitor cells grafted into the grownup epileptic brain restrain seizures in rat fashions of temporal lobe epilepsy. PLoS One. 2014;9(8):e104092. doi: 10.1371/journal.pone.0104092. [DOI] [PMC free article] [PubMed] [Google Scholar]- 35.Kelly S, Bliss TM, Shah AK, et al. Transplanted human fetal neural stem cells survive, migrate, and differentiate in ischemic rat cerebral cortex. Proc Natl Acad Sci U S A. 2004;101(32):11839-11844. doi: 10.1073/pnas.0404474101. [DOI] [PMC free article] [PubMed] [Google Scholar]- 36.Freed CR, Greene PE, Breeze RE, et al. Transplantation of embryonic dopamine neurons for extreme Parkinson’s disease. N Engl J Med. 2001;344(10):710-719. doi: 10.1056/NEJM200103083441002. [DOI] [PubMed] [Google Scholar]- 37.Bradstreet JJ, Sych N, Antonucci N, et al. Efficacy of fetal stem cell transplantation in autism spectrum disorders: an open-labeled pilot study. Cell Transplant. 2014;23(1 Suppl):S105-S112. doi: 10.3727/096368914X684916. [DOI] [PubMed] [Google Scholar]- 38.Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for outlining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy place assertion. Cytotherapy. 2006;8(4):315-317. doi: 10.1080/14653240600855905. [DOI] [PubMed] [Google Scholar]- 39.Giordano A, Galderisi U, Marino IR. From the laboratory bench to the patient’s bedside: an update on clinical trials with mesenchymal stem cells. J Cell Physiol. 2007;211(1):27-35. doi: 10.1002/jcp.20959. [DOI] [PubMed] [Google Scholar]- 40.Katuchova J, Harvanova D, Spakova T, et al. Mesenchymal stem cells in the treatment of kind 1 diabetes mellitus. Endocr Pathol. 2015;26(2):95-103. doi: 10.1007/s12022-015-9362-y. [DOI] [PubMed] [Google Scholar]- 41.Siniscalco D, Bradstreet JJ, Sych N, Antonucci N. Mesenchymal stem cells in treating autism: Novel insights. World J Stem Cells. 2014;6(2):173-178. doi: 10.4252/wjsc.v6.i2.173. [DOI] [PMC free article] [PubMed] [Google Scholar]- 42.Liang CM, Weng SJ, Tsai TH, et al. Neurotrophic and neuroprotective potential of human limbus-derived mesenchymal stromal cells. Cytotherapy. 2014;16(10):1371-1383. doi: 10.1016/j.jcyt.2014.05.015. [DOI] [PubMed] [Google Scholar]- 43.Klyushnenkova E, Mosca JD, Zernetkina V, et al. T cell responses to allogeneic human mesenchymal stem cells: immunogenicity, tolerance, and suppression. J Biomed Sci. 2005;12(1):47-57. doi: 10.1007/s11373-004-8183-7. [DOI] [PubMed] [Google Scholar]- 44.Jacobs SA, Roobrouck VD, Verfaillie CM, Van Gool SW. Immunological characteristics of human mesenchymal stem cells and multipotent adult progenitor cells. Immunol Cell Biol. 2013;91(1):32-39. doi: 10.1038/icb.2012.64. [DOI] [PMC free article] [PubMed] [Google Scholar]- 45.Siniscalco D, Sapone A, Cirillo A, Giordano C, Maione S, Antonucci N. Autism spectrum disorders: is mesenchymal stem cell personalized therapy the long run? J Biomed Biotechnol. 2012;2012:480289. doi: 10.1155/2012/480289. [DOI] [PMC free article] [PubMed] [Google Scholar]- 46.Kassis I, Vaknin-Dembinsky A, Karussis D. Bone marrow mesenchymal stem cells: brokers of immunomodulation and neuroprotection. Curr Stem Cell Res Ther. 2011;6(1):63-68. doi: 10.2174/157488811794480762. [DOI] [PubMed] [Google Scholar]- 47.Karussis D, Petrou P, Kassis I. Clinical experience with stem cells and other cell therapies in neurological diseases. J Neurol Sci. 2013;324(1-2):1-9. doi: 10.1016/j.jns.2012.09.031. [DOI] [PubMed] [Google Scholar]- 48.Lv YT, Zhang Y, Liu M, et al. Transplantation of human cord blood mononuclear cells and umbilical cord-derived mesenchymal stem cells in autism. J Transl Med. 2013;11:196. doi: 10.1186/1479-5876-11-196. [DOI] [PMC free article] [PubMed] [Google Scholar]- 49.Bourin P, Bunnell BA, Casteilla L, et al. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: a joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT) Cytotherapy. 2013;15(6):641-648. doi: 10.1016/j.jcyt.2013.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]- 50.Frese L, Dijkman PE, Hoerstrup SP. Adipose tissue-derived stem cells in regenerative medication. Transfus Med Hemother. 2016;43(4):268-274. doi: 10.1159/000448180. [DOI] [PMC free article] [PubMed] [Google Scholar]- 51.Zhou F, Gao S, Wang L, et al. Human adipose-derived stem cells partially rescue the stroke syndromes by promoting spatial learning and memory in mouse center cerebral artery occlusion model. Stem Cell Res Ther. 2015;6:92. doi: 10.1186/s13287-015-0078-1. [DOI] [PMC free article] [PubMed] [Google Scholar]- 52.Ding DC, Chang YH, Shyu WC, Lin SZ. Human umbilical cord mesenchymal stem cells: a new era for stem cell therapy. Cell Transplant. 2015;24(3):339-347. doi: 10.3727/096368915X686841. [DOI] [PubMed] [Google Scholar]- 53.Xu Y, Huang S, Ma K, Fu X, Han W, Sheng Z. Promising new potential for mesenchymal stem cells derived from human umbilical cord Wharton’s jelly: sweat gland cell-like differentiative capacity. J Tissue Eng Regen Med. 2012;6(8):645-654. doi: 10.1002/term.468. [DOI] [PubMed] [Google Scholar]- 54.Alessio N, Pipino C, Mandatori D, et al. Mesenchymal stromal cells from amniotic fluid are much less prone to senescence compared to those obtained from bone marrow: an in vitro study. J Cell Physiol. 2018;233(11):8996-9006. doi: 10.1002/jcp.26845. [DOI] [PubMed] [Google Scholar]- 55.Witkowska-Zimny M, Wrobel E. Perinatal sources of mesenchymal stem cells: Wharton’s jelly, amnion and chorion. Cell Mol Biol Lett. 2011;16(3):493-514. doi: 10.2478/s11658-011-0019-7. [DOI] [PMC free article] [PubMed] [Google Scholar]- 56.Stevens HE, Smith KM, Rash BG, Vaccarino FM. Neural stem cell regulation, fibroblast growth elements, and the developmental origins of neuropsychiatric disorders. Front Neurosci. 2010;4:59. doi: 10.3389/fnins.2010.00059. [DOI] [PMC free article] [PubMed] [Google Scholar]- 57.Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science. 1992;255(5052):1707-1710. doi: 10.1126/science.1553558. [DOI] [PubMed] [Google Scholar]- 58.Palmer TD, Ray J, Gage FH. FGF-2-responsive neuronal progenitors reside in proliferative and quiescent regions of the grownup rodent brain. Mol Cell Neurosci. 1995;6(5):474-486. doi: 10.1006/mcne.1995.1035. [DOI] [PubMed] [Google Scholar]- 59.Buxbaum JD. Multiple uncommon variants within the etiology of autism spectrum disorders. Dialogues Clin Neurosci. 2009;11(1):35-43. doi: 10.31887/DCNS.2009.11.1/jdbuxbaum. [DOI] [PMC free article] [PubMed] [Google Scholar]- 60.Hsu YC, Lee DC, Chiu IM. Neural stem cells, neural progenitors, and neurotrophic factors. Cell Transplant. 2007;16(2):133-150. [PubMed] [Google Scholar]- 61.Kim SU, de Vellis J. Stem cell-based mostly cell therapy in neurological diseases: a evaluate. J Neurosci Res. 2009;87(10):2183-2200. doi: 10.1002/jnr.22054. [DOI] [PubMed] [Google Scholar]- 62.Liu SP, Fu RH, Huang SJ, et al. Stem cell applications in regenerative medication for neurological disorders. Cell Transplant. 2013;22(4):631-637. doi: 10.3727/096368912X655145. [DOI] [PubMed] [Google Scholar]- 63.Siniscalco D, Bradstreet JJ, Antonucci N. Therapeutic function of hematopoietic stem cells in autism spectrum disorder-related inflammation. Front Immunol. 2013;4:140. doi: 10.3389/fimmu.2013.00140. [DOI] [PMC free article] [PubMed] [Google Scholar]- 64.Brigida AL, Siniscalco D. Induced pluripotent stem cells as a cellular model for studying Down Syndrome. J Stem Cells Regen Med. 2016;12(2):54-60. doi: 10.46582/jsrm.1202009. [DOI] [PMC free article] [PubMed] [Google Scholar]- 65.Park IH, Arora N, Huo H, et al. Disease-particular induced pluripotent stem cells. Cell. 2008;134(5):877-886. doi: 10.1016/j.cell.2008.07.041. [DOI] [PMC free article] [PubMed] [Google Scholar]- 66.Medvedev SP, Shevchenko AI, Zakian SM. Induced pluripotent stem cells: problems and advantages when applying them in regenerative medication. Acta Naturae. 2010;2(2):18-28. [PMC free article] [PubMed] [Google Scholar]- 67.Nori S, Okada Y, Yasuda A, et al. Grafted human-induced pluripotent stem-cell-derived neurospheres promote motor useful restoration after spinal cord injury in mice. Proc Natl Acad Sci U S A. 2011;108(40):16825-16830. doi: 10.1073/pnas.1108077108. [DOI] [PMC free article] [PubMed] [Google Scholar]- 68.Hallett PJ, Deleidi M, Astradsson A, et al. Successful operate of autologous iPSC-derived dopamine neurons following transplantation in a non-human primate mannequin of Parkinson’s illness. Cell Stem Cell. 2015;16(3):269-274. doi: 10.1016/j.stem.2015.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]- 69.Jeon I, Choi C, Lee N, et al. In vivo roles of a patient-derived induced pluripotent stem cell line (HD72-iPSC) within the YAC128 model of Huntington’s illness. Int J Stem Cells. 2014;7(1):43-47. doi: 10.15283/ijsc.2014.7.1.43. [DOI] [PMC free article] [PubMed] [Google Scholar]- 70.Nizzardo M, Simone C, Rizzo F, et al. Minimally invasive transplantation of iPSC-derived ALDHhiSSCloVLA4+ neural stem cells successfully improves the phenotype of an amyotrophic lateral sclerosis model. Hum Mol Genet. 2014;23(2):342-354. doi: 10.1093/hmg/ddt425. [DOI] [PMC free article] [PubMed] [Google Scholar]- 71.Knoepfler PS. Deconstructing stem cell tumorigenicity: a roadmap to secure regenerative drugs. Stem Cells. 2009;27(5):1050-1056. doi: 10.1002/stem.37. [DOI] [PMC free article] [PubMed] [Google Scholar]- 72.Zhao T, Zhang ZN, Rong Z, Xu Y. Immunogenicity of induced pluripotent stem cells. Nature. 2011;474(7350):212-215. doi: 10.1038/nature10135. [DOI] [PubMed] [Google Scholar]- 73.Ben-Reuven L, Reiner O. Modeling the autistic cell: iPSCs recapitulate developmental rules of syndromic and nonsyndromic ASD. Dev Growth Differ. 2016;58(5):481-491. doi: 10.1111/dgd.12280. [DOI] [PubMed] [Google Scholar]- 74.Varga E, Nemes C, Bock I, et al. Establishment of an induced pluripotent stem cell (iPSC) line from a 9-yr previous male with autism spectrum disorder (ASD) Stem Cell Res. 2017;21:19-22. doi: 10.1016/j.scr.2017.03.013. [DOI] [PubMed] [Google Scholar]- 75.Lim CS, Yang JE, Lee YK, Lee K, Lee JA, Kaang BK. Understanding the molecular basis of autism in a dish using hiPSCs-derived neurons from ASD patients. Mol Brain. 2015;8(1):57. doi: 10.1186/s13041-015-0146-6. [DOI] [PMC free article] [PubMed] [Google Scholar]- 76.Kim DS, Ross PJ, Zaslavsky K, Ellis J. Optimizing neuronal differentiation from induced pluripotent stem cells to model ASD. Front Cell Neurosci. 2014;8:109. doi: 10.3389/fncel.2014.00109. [DOI] [PMC free article] [PubMed] [Google Scholar]- 77.Griesi-Oliveira K, Acab A, Gupta AR, et al. Modeling non-syndromic autism and the affect of TRPC6 disruption in human neurons. Mol Psychiatry. 2015;20(11):1350-1365. doi: 10.1038/mp.2014.141. [DOI] [PMC free article] [PubMed] [Google Scholar]- 78.Liu X, Campanac E, Cheung HH, et al. Idiopathic autism: cellular and molecular phenotypes in pluripotent stem cell-derived neurons. Mol Neurobiol. 2017;54(6):4507-4523. doi: 10.1007/s12035-016-9961-8. [DOI] [PMC free article] [PubMed] [Google Scholar]- 79.Kathuria A, Nowosiad P, Jagasia R, et al. Stem cell-derived neurons from autistic individuals with SHANK3 mutation present morphogenetic abnormalities during early growth. Mol Psychiatry. 2018;23(3):735-746. doi: 10.1038/mp.2017.185. [DOI] [PMC free article] [PubMed] [Google Scholar]- 80.Nestor MW, Phillips AW, Artimovich E, Nestor JE, Hussman JP, Blatt GJ. Human inducible pluripotent stem cells and autism spectrum disorder: emerging technologies. Autism Res. 2016;9(5):513-535. doi: 10.1002/aur.1570. [DOI] [PubMed] [Google Scholar]- 81.Tania M, Khan MA, Xia K. Recent advances in animal model experimentation in autism analysis. Acta Neuropsychiatr. 2014;26(5):264-271. doi: 10.1017/neu.2013.58. [DOI] [PubMed] [Google Scholar]- 82.Segal-Gavish H, Karvat G, Barak N, et al. Mesenchymal stem cell transplantation promotes neurogenesis and ameliorates autism related behaviors in BTBR mice. Autism Res. 2016;9(1):17-32. doi: 10.1002/aur.1530. [DOI] [PubMed] [Google Scholar]- 83.Perets N, Segal-Gavish H, Gothelf Y, et al. Long run helpful impact of neurotrophic factors-secreting mesenchymal stem cells transplantation in the BTBR mouse mannequin of autism. Behav Brain Res. 2017;331:254-260. doi: 10.1016/j.bbr.2017.03.047. [DOI] [PubMed] [Google Scholar]- 84.Ha S, Park H, Mahmood U, Ra JC, Suh YH, Chang KA. Human adipose-derived stem cells ameliorate repetitive habits, social deficit and anxiety in a VPA-induced autism mouse mannequin. Behav Brain Res. 2017;317:479-484. doi: 10.1016/j.bbr.2016.10.004. [DOI] [PubMed] [Google Scholar]- 85.Gobshtis N, Tfilin M, Wolfson M, Fraifeld VE, Turgeman G. Transplantation of mesenchymal stem cells reverses behavioural deficits and impaired neurogenesis attributable to prenatal publicity to valproic acid. Oncotarget. 2017;8(11):17443-17452. doi: 10.18632/oncotarget.15245. [DOI] [PMC free article] [PubMed] [Google Scholar]- 86.Hsiao EY, McBride SW, Chow J, Mazmanian SK, Patterson PH. Modeling an autism danger think about mice leads to everlasting immune dysregulation. Proc Natl Acad Sci U S A. 2012;109(31):12776-12781. doi: 10.1073/pnas.1202556109. [DOI] [PMC free article] [PubMed] [Google Scholar]- 87.Griesi-Oliveira K, Sunaga DY, Alvizi L, Vadasz E, Passos-Bueno MR. Stem cells as a good software to research dysregulated biological systems in autism spectrum disorders. Autism Res. 2013;6(5):354-361. doi: 10.1002/aur.1296. [DOI] [PubMed] [Google Scholar]- 88.Sharma A, Gokulchandran N, Sane H, et al. Autologous bone marrow mononuclear cell therapy for autism: an open label proof of concept examine. Stem Cells Int. 2013;2013:623875. doi: 10.1155/2013/623875. [DOI] [PMC free article] [PubMed] [Google Scholar]- 89.Bjørklund G, Kern JK, Urbina MA, et al. Cerebral hypoperfusion in autism spectrum disorder. Acta Neurobiol Exp (Wars) 2018;78(1):21-29. [PubMed] [Google Scholar]- 90.Sharma A, Gokulchandran N, Sane H, et al. Therapeutic effects of cellular therapy in a case of adult autism spectrum of disorder. Int Biol Biomed J. 2018;4(2):98-103. [Google Scholar]- 91.Gao S, Liu F. Fetal liver: an ideal niche for hematopoietic stem cell enlargement. Sci China Life Sci. 2018;61(8):885-892. doi: 10.1007/s11427-018-9313-4. [DOI] [PubMed] [Google Scholar]- 92.Chez M, Lepage C, Parise C, Dang-Chu A, Hankins A, Carroll M. Safety and observations from a placebo-managed, crossover study to assess use of autologous umbilical cord blood stem cells to improve signs in children with autism. Stem Cells Transl Med. 2018;7(4):333-341. doi: 10.1002/sctm.17-0042. [DOI] [PMC free article] [PubMed] [Google Scholar]- 93.Dawson G, Sun JM, Davlantis KS, et al. Autologous cord blood infusions are secure and possible in younger children with autism spectrum disorder: outcomes of a single-middle Phase I open-label trial. Stem Cells Transl Med. 2017;6(5):1332-1339. doi: 10.1002/sctm.16-0474. [DOI] [PMC free article] [PubMed] [Google Scholar]- 94.Siniscalco D, Pandolfi A, Galderisi U. State-of-the-artwork on basic and utilized stem cell therapy; Stem Cell Research Italy-International Society for Cellular Therapy Europe. Stem Cells Dev. 2012;21(5):668-669. doi: 10.1089/scd.2011.0609. [DOI] [PubMed] [Google Scholar]- 95.Huang H, Young W, Chen L, et al. Clinical cell therapy tips for neurorestoration (IANR/CANR 2017) Cell Transplant. 2018;27(2):310-324. doi: 10.1177/0963689717746999. [DOI] [PMC free article] [PubMed] [Google Scholar]- 96.Siniscalco D, Giordano A, Galderisi U. Novel insights in primary and utilized stem cell therapy. J Cell Physiol. 2012;227(5):2283-2286. doi: 10.1002/jcp.22945. [DOI] [PubMed] [Google Scholar]- 97.Squillaro T, Peluso G, Galderisi U. Clinical trials with mesenchymal stem cells: an update. Cell Transplant. 2016;25(5):829-848. doi: 10.3727/096368915X689622. [DOI] [PubMed] [Google Scholar]- 98.Harousseau JL. Role of stem cell transplantation. Hematol Oncol Clin North Am. 2007;21(6):1157-1174x. doi: 10.1016/j.hoc.2007.08.001. [DOI] [PubMed] [Google Scholar]- 99.Steward CG, Jarisch A. Haemopoietic stem cell transplantation for genetic disorders. Arch Dis Child. 2005;90(12):1259-1263. doi: 10.1136/adc.2005.074278. [DOI] [PMC free article] [PubMed] [Google Scholar]- 100.Caglayan AO. Genetic causes of syndromic and non-syndromic autism. Dev Med Child Neurol. 2010;52(2):130-138. doi: 10.1111/j.1469-8749.2009.03523.x. [DOI] [PubMed] [Google Scholar]- 101.Sharma A, Gokulchandran N, Chopra G, et al. Administration of autologous bone marrow-derived mononuclear cells in kids with incurable neurological disorders and damage is safe and improves their quality of life. Cell Transplant. 2012;21(Suppl 1):S79-S90. doi: 10.3727/096368912X633798. [DOI] [PubMed] [Google Scholar]- 102.Siniscalco D, Di Marsilio A, Antonucci N. Ethics in autism care. Autism. 2013;3:e119. [Google Scholar]- 103.Hess DC, Borlongan CV. Stem cells and neurological diseases. Cell Prolif. 2008;41(Suppl 1):94-114. doi: 10.1111/j.1365-2184.2008.00486.x. [DOI] [PMC free article] [PubMed] [Google Scholar]- 104.Siniscalco D, Bradstreet JJ, Antonucci N. The promise of regenerative medication and stem cell analysis for the therapy of autism. . J Regen Med.

If you have any sort of inquiries concerning where and how you can use https://autismtreatmentcare.com/, you could contact us at our own webpage.

Personal tools