Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Stem Cells. Apr 26, 2014; 6(2): 173-178
Published online Apr 26, 2014. doi: 10.4252/wjsc.v6.i2.173
Mesenchymal stem cells in treating autism: Novel insights
Dario Siniscalco, Department of Experimental Medicine, Second University of Naples, 16-80138 Napoli, Italy
Dario Siniscalco, Centre for Autism, La Forza del Silenzio, 81036 Caserta, Italy
Dario Siniscalco, Cancellautismo, 50132 Florence, Italy
James Jeffrey Bradstreet, Brain Treatment Center, Buford, GA 30518, United States
Nataliia Sych, Clinical Department, Cell Therapy Center EmCell, 04073 Kiev, Ukraine
Nicola Antonucci, Biomedical Centre for Autism Research and Treatment, 70126 Bari, Italy
Author contributions: Siniscalco D designed the paper; Siniscalco D and Bradstreet JJ wrote the paper; Sych N contributed to the paragraph on MSCs; Antonucci N contributed to the paragraph on autism; Bradstreet JJ edited the English language.
Correspondence to: Dario Siniscalco, ChemD, PhD, Department of Experimental Medicine, Second University of Naples, Via S Maria di Costantinopoli, 16-80138 Napoli, Italy. dariosin@uab.edu
Telephone: +39-81-5665880 Fax: +39-81-5667503
Received: November 11, 2013
Revised: December 19, 2013
Accepted: March 17, 2014
Published online: April 26, 2014

Abstract

Autism spectrum disorders (ASDs) are complex neurodevelopmental disorders characterized by dysfunctions in social interactions, abnormal to absent verbal communication, restricted interests, and repetitive stereotypic verbal and non-verbal behaviors, influencing the ability to relate to and communicate. The core symptoms of ASDs concern the cognitive, emotional, and neurobehavioural domains. The prevalence of autism appears to be increasing at an alarming rate, yet there is a lack of effective and definitive pharmacological options. This has created an increased sense of urgency, and the need to identify novel therapies. Given the growing awareness of immune dysregulation in a significant portion of the autistic population, cell therapies have been proposed and applied to ASDs. In particular, mesenchymal stem cells (MSCs) possess the immunological properties which make them promising candidates in regenerative medicine. MSC therapy may be applicable to several diseases associated with inflammation and tissue damage, where subsequent regeneration and repair is necessary. MSCs could exert a positive effect in ASDs through the following mechanisms: stimulation of repair in the damaged tissue, e.g., inflammatory bowel disease; synthesizing and releasing anti-inflammatory cytokines and survival-promoting growth factors; integrating into existing neural and synaptic network, and restoring plasticity. The paracrine mechanisms of MSCs show interesting potential in ASD treatment. Promising and impressive results have been reported from the few clinical studies published to date, although the exact mechanisms of action of MSCs in ASDs to restore functions are still largely unknown. The potential role of MSCs in mediating ASD recovery is discussed in light of the newest findings from recent clinical studies.

Key Words: Autism spectrum disorders, Autism treatment, Cell therapy, Mesenchymal stem cells

Core tip: Autism spectrum disorders are still untreatable pathologies. Mesenchymal stem cells possess the immunological properties which make them promising candidates as a novel therapeutic option.



AUTISM SPECTRUM DISORDERS

Autism spectrum disorders (ASDs) are complex neurodevelopmental disorders. Indeed, this term refers to a heterogenous group of varied conditions characterized by dysfunctions in social interactions, skills, and communication, restricted interests, and repetitive stereotypic verbal and non-verbal behaviors, influencing the ability to relate to others. Cognitive, emotional and neurobehavioral abnormalities characterize the core symptoms[1,2]. The prevalence of these disorders has dramatically increased in the last years, with present rates of 11.3 per 1000 (one in 88) children aged 8 years in the United States, according to Centers for Disease Control[3]. ASDs are presumed to be a lifelong disability with multiple impacts on child and adult health. Indeed, adult autistic individuals show limited independence because of their learning disability. In adulthood, communication is still impaired, as reading and spelling abilities are poor. Stereotyped behaviors and restricted interests still persist. The children affected require special and intensive parental, school, and social support[4]. ASD results in a substantial impact on a person’s quality of life and that of their family[5]. Given the total lifetime societal cost of caring for one individual with autism, estimated in $3.2 million[6], autism should be considered as an urgent public health priority[2].

Together with the cognitive, emotional and neurobehavioral abnormalities, ASDs are disorders characterized by a broad range of biochemical, toxicological and immune involvement, including: oxidative stress, endoplasmic reticulum stress, decreased methylation capacity, limited production of glutathione, mitochondrial dysfunction, intestinal dysbiosis, increased toxic metal burden, and immune dysregulations including autoimmunity[7].

Currently, only a handful of medications are licensed for treating a limited number of autism-related symptoms[8]. Moreover, prescribed pharmaceuticals (i.e., anti-psychotics) fail to address the ASD core symptoms, have the potential of markedly adverse effects, and are at best palliative[9-12]. The alternative treatments for ASDs are diverse and include: behavioral, nutritional, and biomedical approaches. Thus the need for a definitive and effective therapy is an unfulfilled priority for autism research.

MESENCHYMAL STEM CELLS

Presently, cell therapies and cell-based biopharmacies offer a valid intervention for several otherwise untreatable human diseases. Stem cells appear to represent the greatest potential for the future of molecular and regenerative medicine[13,14]. Among the various stem cell subtypes, mesenchymal stem cells (MSCs) provide a useful tool for the treatment of several diseases associated with inflammation, tissue damage, and subsequent regeneration and repair[15].

MSCs are multipotent stem cells that posses the capacity to differentiate in vivo or in vitro, under specific conditions, into cells of connective tissue lineages, including bone, fat, cartilage and muscle[16]. They are distinct from the hematopoietic lineage, and were initially described by Alexander Friedenstein in the 1960s after he extracted MSCs from bone marrow[17]. It is common practice for clinical and research applications, to acquire MSCs from bone marrow aspirates of the superior iliac crest under local anesthesia. The cells are then isolated by their adherence to plastic and amplified through passage in culture, where they exhibit a great replicative capacity[18].

In order to achieve a detailed classification of this type of stem cell, the International Society for Cellular Therapy has proposed the following minimal criteria to identify human MSCs: they must grow in standard, plastic-adherent culture conditions; must express the cyto-specific markers CD73, CD90 and CD105, without expression of CD45, CD34, CD14 or CD11b, CD79alpha or CD19 and HLA-DR surface molecules; and must be capable of in vitro differentiation into osteoblasts, adipocytes and chondroblasts[19].

Interestingly, MSCs seem to be the most promising clinical candidate for immune-modulatory cell-based therapy[20]. MSCs show immunomodulatory capacities, as they are able to induce tolerance in immunocompetent allotransplants or even xenotransplant recipients[21]. Interacting with a wide range of immune cells, probably through a cell-to-cell contact mechanism[22], MSCs are able to modulate T-cell phenotype and immune-suppress the local environment[23].

Their unique properties of immunomodulation, multipotency, and rapid self-renewal proliferation rate, distinguish them as useful tools for application in immunomodulatory therapy and neurological disorders. In addition, other desirable characteristics of MSCs, e.g., genetic stability, stable phenotype, and easy procedures for collection, storage and shipping from the laboratory to the bedside[24], direct us to MSC-based therapies as a potent intervention.

In clinical settings, MSCs can be transplanted directly without genetic modification or pretreatments (i.e., immunosuppressants). No host vs graft rejection has been observed[25]. Importantly, there is an absence of uncontrollable growth or tumorigenesis with MSCs, in contrast to the potential problems intrinsic to embryonic stem cells[26]. Crucially, MSCs create no moral objection or ethical-religious controversies, unlike embryonic or fetal stem cells[27].

MESENCHYMAL STEM CELLS IN TREATING AUTISM: THE RATIONALE

The potential application of cell therapy, in particular MSCs, for ASDs has already been discussed by our group[28,29]. After a brief description of MSC-mediated ameliorative effects in ASDs, we will review recent and ongoing clinical trials using MSC transplantation in ASD patients.

We hypothesize that MSCs exert a positive effect in ASDs through the following mechanisms: stimulation of the plastic response in the host damaged tissue (e.g., inflammatory bowel disorders); synthesizing and releasing anti-inflammatory cytokines and survival-promoting growth factors (paracrine and biopharmacy); integrating into existing neural and synaptic network (engrafting), and restoring plasticity[28,29]. Following transplantation, MSCs target and migrate to the site of injury. In some cases these cells respond to the local environment with appropriate secretion of soluble factors to ameliorate inflammation and promote repair[30]. This paracrine mechanism offers potential in ASD treatment.

ASDs are characterized by a coexistent, if not etiological, immune system dysregulation[31]. Changes in innate and adaptive immune responses have been reported in ASD patients[32]. Characteristically, ASD cases show alterations in both T cell- and B cell-mediated immunity, as well as an imbalance in CD3+, CD4+, and CD8+ T cells and natural killer (NK) cells[33]. On these bases, the regulatory effects mediated by MSCs present an optimal way to restore immune balance, which cannot otherwise be obtained through pharmaceutical interventions. Through inhibition of the proliferation of CD8+/CD4+ T lymphocytes and NK cells, suppression of the immunoglobulin production by plasma cells, and inhibition of the maturation of dendritic cells (DCs), MSC transplantation appears ideally suited to provide a unique therapeutic application for ASDs[34,35].

In addition, MSCs are able to inhibit T lymphocyte pro-inflammatory cytokine production[36]. MSCs function as an implanted biopharmacy: after homing in to the targeted tissue site, they synthesize and release a broad range of bioactive molecules[35,37], i.e., anti-inflammatory cytokines, trophic and growth factors, interleukin (IL)-6, IL-7, IL-8, IL-11, IL-12, IL-14, IL-15, macrophage colony-stimulating factor, Flt-3 ligand, and stem-cell factor[38], which in turn could be responsible for activating endogenous restorative mechanisms within injured tissues. This strong paracrine activity of MSCs seems to be the most plausible and reasonable mechanism for the functional benefit derived from MSC transplantation. Furthermore, transplanted MSCs can induce the host tissue to upregulate the production of anti-inflammatory molecules, such as IL-10, in this way restoring the pro-inflammatory processes noted in ASDs[39,40].

MESENCHYMAL STEM CELLS IN TREATING AUTISM: CLINICAL EVIDENCE

Despite insufficient pre-clinical models of MSC therapy for ASDs[41], several clinical studies on humans have been conducted. Recently, a non-randomized, open-label, controlled, single-center phase I/II clinical trial to examine the treatment safety and efficacy of transplantation of human cord blood mononuclear cell (CBMNCs) and/or human umbilical cord-derived mesenchymal stem cells (UCMSCs) in children with autism has been performed[42]. Stem cell administration was carried out via intravenous and intrathecal infusions. Autistic children transplanted with cells were followed for 24 wk. According to the authors, the cell treatment was safe, well tolerated without immediate or long term side effects, and no allergic, immunological reactions or other serious adverse events were observed at the time of injection or during the whole follow-up period. The cell transplantation showed efficacy; improvements were observed in visual, emotional and intellectual responses, body use, adaption to change, fear or nervousness, non-verbal communication and activity level, as measured by Childhood Autism Rating Scale, as well as in lethargy/social withdrawal, stereotypic behavior, hyperactivity and inappropriate speech evaluated by the Aberrant Behavior Checklist[42]. They noted that the group receiving CBMNCs and UCMNCs demonstrated a more robust therapeutic effect than the group receiving mono-cell line therapy, which may be attributed to the action of CBMNCs and UCMSCs in synergy. It has been proposed the synergistic mechanism is related to increased cell-mediated perfusion in brain areas and/or the regulation of immune dysfunction.

Intrathecally transplanted autologous bone marrow-derived mononuclear cells were efficacious in improving the quality of life in a 14-year-old boy with severe autism[43]. A detailed cell-sorting analysis was not done, but the cell extract contained a percentage of MSCs. We know bone marrow is comprised of a heterogeneous population of stem cells, encompassing hematopoietic stem cells, MSCs, endothelial progenitor cells, and very small embryonic-like stem cells. The bone marrow cell transplantation was safe, the patient had no noted side-effects and showed some immediate improvements within a week (eye contact and attention, fine motor activities). Significant improvements were observed over a period of 6 mo to 1 year (social interaction and emotions, impulse control, reading skills, tracing, recognition of all shapes and following commands, and hyperactivity). Interestingly, comparisons of pre/post cell therapy brain positron emission tomography scans revealed a markedly increased uptake in bilateral temporal lobes and bilateral calcarine cortices with mild increased uptake in the left medial pre-frontal cortex[43].

Transplanted stem cells therefore seemed to ameliorate neural hypoperfusion in the previous case report. Hypoperfusion may be a consequence of focal inflammation and would likely result in low-grade ischemic consequences: hypoxia, abnormal metabolites, neurotransmitters dysregulation, and potential neural tissue damage.

In the light of these encouraging, but limited observations, the authors launched an open-label proof-of-concept study using autologous bone marrow-derived mononuclear cell transplantation in 32 patients with autism[44]. The average number of intrathecally injected cells was 8 × 107 cells. Cell treatment was determined to be safe and adverse events were transient (hyperactivity). They hypothesize that the intrathecal administration route is able to enhance homing of the transplanted cells into the central nervous system. Clinical improvements after cellular therapy were observed in social relationships and reciprocity, emotional responsiveness, communication and behavior. As a putative mechanism of action, the authors further hypothesized that cellular transplantation was able to restore function to ASD patients by neuroprotection, neural circuit reconstruction, neural plasticity, neurogenesis, and immunomodulation.

The hypothesis that intrathecal administration increases the efficacy of stem cell therapies is not actually evaluated by these various studies. Clearly, it is a testable hypothesis and future studies should include arms with and without intrathecal administration to compare the therapeutic efficacy of the more invasive intrathecal implantations.

PROBLEMS

Despite these early clinical trials with MSCs, there are no apparent pre-clinical studies on the use of MSCs in ASD models[41]. Thus, more research into the mechanisms of action post transplantation is required to adequately understand the route, dosing and safety. However, the parental perspective is unlikely to wait on more detailed scientific studies. Stem cells are readily available from many centers in numerous countries, with various cell types and methodologies being utilized. Families recognize the devastating nature of autism on their children and are already seeking stem cell therapies. Based on a simple scan of the internet sites advertising cell therapies, it appears hundreds of ASD children have already been treated.

Another complexity in the research arises from stem cell sourcing. Some protocols use allogeneic (derived from a different person or collection of donors), while others use autologous donor (self-derived) cell types[15]. Some protocols for ASD also use expanded autologous MSCs (United States Patent Application: 20060182724). This adds another dimension to the discussion and a potential source of laboratory contamination. Expansion requires medium for growth from which the cells must then be isolated, and any medium washed sufficiently to prevent a reaction in the recipient. Typically bovine serum is used. This creates the further risk of prion infection of the medium. To avoid this, one group has proposed using pooled human serum[45]. This xeno-free methodology has many desirable features, but retains the concerns about human pathogen transmission. The group, however, screened extensively for contamination and it appeared they were able to ascertain that the samples were free of any disease vectors. This process should be considered for any use of expanded MSCs for ASD therapies.

Another challenge in the standardization of dosing derives from the varying efficacy amongst allogeneic donors in terms of: vitality, potency, and expansion potential. Every donor is different and this could affect efficacy and also paracrine effects. Indeed, it seems that the secretion of bioactive molecules could differ by a factor of 10 between different donors of matched age and gender[15]. Recently, in order to increase the adequate supply of stem cells from donor tissues, it has been demonstrated that a 3D co-culture system with murine-derived hematopoietic stem cells co-cultured with MSCs produces 3D-microaggregates of stem cells. These 3D-microaggregate systems support the expansion of approximately twice as many hematopoietic stem cell candidates as the 2D controls. In addition, the MSCs maintained in 3D aggregates are able to express significantly higher levels of hematopoietic niche factors compared with 2D cultures[46].

Finally, there are complex hurdles to overcome from the legal and regulatory restrictions placed by governments seeking to control cell therapies[27]. Several countries (i.e., United States and EU area) have attempted to create uniformity within the regulations governing cell trials, while creating very stringent regulations on cell culture conditions, diseases to be treated, and patient safety. However, in some other countries (e.g., Ukraine, China, Dominican Republic, Panama, and Mexico) the access to cell therapy is more readily available.

CONCLUSION

The rapidly increasing prevalence of ASDs worldwide is creating an urgent need for effective restorative therapies. The lack of safe and effective psychopharmaceuticals and other definitive medical therapies, together with the limited understanding of the pathophysiology, has created an urgency to identify novel and more effective therapies[47]. MSCs appear to offer a greater potential in regenerative medicine for complex disorder like autism than existing pharmaceutical protocols. Promising and impressive early results have been achieved from a few clinical studies, although the exact restorative mechanisms of action of MSCs in ASDs are still largely unknown.

ACKNOWLEDGMENTS

The authors gratefully thank Mr. Enzo Abate, Ms. Giovanna Gallone, and the nonprofit ozrganizations “La Forza del Silenzio” and “Cancellautismo,” Italy for their useful assistance.

Footnotes

P- Reviewers: Shawcross SG, Tanabe S, Yao CL S- Editor: Gou SX L- Editor: Cant MR E- Editor: Zhang DN

References
1.  American Psychiatric Association Diagnostic and Statistical Manual of Mental Disorders. 4th ed. Washington, DC: American Psychiatric Pub 2000; .  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Siniscalco D. Current Findings and Research Prospective in Autism Spectrum Disorders. Autism. 2013;S2:e001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
3.  Autism and Developmental Disabilities Monitoring Network Surveillance Year 2008 Principal Investigators; Centers for Disease Control and Prevention. Prevalence of autism spectrum disorders--Autism and Developmental Disabilities Monitoring Network, 14 sites, United States, 2008. MMWR Surveill Summ. 2012;61:1-19.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Siniscalco D, Cirillo A, Bradstreet JJ, Antonucci N. Epigenetic findings in autism: new perspectives for therapy. Int J Environ Res Public Health. 2013;10:4261-4273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
5.  Moyal WN, Lord C, Walkup JT. Quality of life in children and adolescents with autism spectrum disorders: what is known about the effects of pharmacotherapy? Paediatr Drugs. 2014;16:123-128.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Randolph-Gips M, Srinivasan P. Modeling autism: a systems biology approach. J Clin Bioinforma. 2012;2:17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
7.  Bradstreet JJ, Smith S, Baral M, Rossignol DA. Biomarker-guided interventions of clinically relevant conditions associated with autism spectrum disorders and attention deficit hyperactivity disorder. Altern Med Rev. 2010;15:15-32.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Siniscalco D, Antonucci N. Involvement of dietary bioactive proteins and peptides in autism spectrum disorders. Curr Protein Pept Sci. 2013;14:674-679.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Siniscalco D, Antonucci N. Possible use of Trichuris suis ova in autism spectrum disorders therapy. Med Hypotheses. 2013;81:1-4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
10.  Chadman KK, Guariglia SR, Yoo JH. New directions in the treatment of autism spectrum disorders from animal model research. Expert Opin Drug Discov. 2012;7:407-416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
11.  Doyle CA, McDougle CJ. Pharmacotherapy to control behavioral symptoms in children with autism. Expert Opin Pharmacother. 2012;13:1615-1629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
12.  Owen-Smith A, Bent S, Lynch F, Coleman K, Yau V, Freiman H, Pearson K, Massolo M, Pomichowski M, Croen L. Complementary and Alternative Medicine Use Among Children with Autism Spectrum Disorders: Findings from the Mental Health Research Network Autism Registry Web Survey. Clin Med Res. 2013;11:163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
13.  Siniscalco D, Giordano A, Galderisi U. Novel insights in basic and applied stem cell therapy. J Cell Physiol. 2012;227:2283-2286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 12]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
14.  Siniscalco D, Pandolfi A, Galderisi U. State-of-the-art on basic and applied stem cell therapy; Stem Cell Research Italy-International Society for Cellular Therapy Europe, Joint Meeting, Montesilvano (PE)-Italy, June 10-12, 2011. Stem Cells Dev. 2012;21:668-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
15.  Dimarino AM, Caplan AI, Bonfield TL. Mesenchymal Stem Cells in Tissue Repair. Front Immunol. 2013;4:201.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol. 2004;36:568-584.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Friedenstein AJ, Gorskaja JF, Kulagina NN. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp Hematol. 1976;4:267-274.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Digirolamo CM, Stokes D, Colter D, Phinney DG, Class R, Prockop DJ. Propagation and senescence of human marrow stromal cells in culture: a simple colony-forming assay identifies samples with the greatest potential to propagate and differentiate. Br J Haematol. 1999;107:275-281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 670]  [Cited by in F6Publishing: 643]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
19.  Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj, Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315-317.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Oh SKW, Choo ABH. The Biological Basis Stem Cells. Comprehensive Biotechnology. 2nd ed. Amsterdam: Elsevier 2011; 341-365.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Atoui R, Chiu RC. Concise review: immunomodulatory properties of mesenchymal stem cells in cellular transplantation: update, controversies, and unknowns. Stem Cells Transl Med. 2012;1:200-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 65]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
22.  Siniscalco D, Giordano C, Galderisi U, Luongo L, de Novellis V, Rossi F, Maione S. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 76]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
23.  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;91:32-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 164]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
24.  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;211:27-35.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Sotiropoulou PA, Papamichail M. Immune properties of mesenchymal stem cells. Methods Mol Biol. 2007;407:225-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 43]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
26.  Siniscalco D, Giordano C, Galderisi U, Luongo L, Alessio N, Di Bernardo G, de Novellis V, Rossi F, Maione S. Intra-brain microinjection of human mesenchymal stem cells decreases allodynia in neuropathic mice. Cell Mol Life Sci. 2010;67:655-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 68]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
27.  Siniscalco D. Suspended Life-Stem Cells: Are Treatments Possible? J Regen Med. 2012;2:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
28.  Siniscalco D, Sapone A, Cirillo A, Giordano C, Maione S, Antonucci N. Autism spectrum disorders: is mesenchymal stem cell personalized therapy the future? J Biomed Biotechnol. 2012;2012:480289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 33]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
29.  Siniscalco D, Bradstreet JJ, Sych N, Antonucci N. Perspectives on the Use of Stem Cells for Autism Treatment. Stem Cells Int. 2013;2013:262438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
30.  Sohni A, Verfaillie CM. Mesenchymal Stem Cells Migration Homing and Tracking. Stem Cells Int. 2013;2013:130763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 277]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
31.  Ashwood P, Corbett BA, Kantor A, Schulman H, Van de Water J, Amaral DG. In search of cellular immunophenotypes in the blood of children with autism. PLoS One. 2011;6:e19299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 80]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
32.  Gupta S, Samra D, Agrawal S. Adaptive and Innate Immune Responses in Autism: Rationale for Therapeutic Use of Intravenous Immunoglobulin. J Clin Immunol. 2010;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Gupta S. Immunological treatments for autism. J Autism Dev Disord. 2000;30:475-479.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Hoogduijn MJ, Popp F, Verbeek R, Masoodi M, Nicolaou A, Baan C, Dahlke MH. The immunomodulatory properties of mesenchymal stem cells and their use for immunotherapy. Int Immunopharmacol. 2010;10:1496-1500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 175]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
35.  Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S, Gianni AM. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood. 2002;99:3838-3843.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Meyerrose T, Olson S, Pontow S, Kalomoiris S, Jung Y, Annett G, Bauer G, Nolta JA. Mesenchymal stem cells for the sustained in vivo delivery of bioactive factors. Adv Drug Deliv Rev. 2010;62:1167-1174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 117]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
37.  Ivanova-Todorova E, Bochev I, Dimitrov R, Belemezova K, Mourdjeva M, Kyurkchiev S, Kinov P, Altankova I, Kyurkchiev D. Conditioned medium from adipose tissue-derived mesenchymal stem cells induces CD4+FOXP3+ cells and increases IL-10 secretion. J Biomed Biotechnol. 2012;2012:295167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
38.  Meirelles Lda S, Fontes AM, Covas DT, Caplan AI. Mechanisms involved in the therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor Rev. 2009;20:419-427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 951]  [Cited by in F6Publishing: 947]  [Article Influence: 63.1]  [Reference Citation Analysis (0)]
39.  Siniscalco D, Sapone A, Giordano C, Cirillo A, de Novellis V, de Magistris L, Rossi F, Fasano A, Maione S, Antonucci N. The expression of caspases is enhanced in peripheral blood mononuclear cells of autism spectrum disorder patients. J Autism Dev Disord. 2012;42:1403-1410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 42]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
40.  El-Ansary A, Al-Ayadhi L. Neuroinflammation in autism spectrum disorders. J Neuroinflammation. 2012;9:265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 96]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
41.  Siniscalco D, Bradstreet JJ, Antonucci N. Therapeutic role of hematopoietic stem cells in autism spectrum disorder-related inflammation. Front Immunol. 2013;4:140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
42.  Lv YT, Zhang Y, Liu M, Qiuwaxi JN, Ashwood P, Cho SC, Huan Y, Ge RC, Chen XW, Wang ZJ. Transplantation of human cord blood mononuclear cells and umbilical cord-derived mesenchymal stem cells in autism. J Transl Med. 2013;11:196.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Sharma A, Badhe P, Gokulchandran N, Kulkarni P, Mishra P, Shetty A, Sane H. An Improved Case of Autism as Revealed by PET CT Scan in Patient Transplanted with Autologous Bone Marrow Derived Mononuclear Cells. J Stem Cell Res Ther. 2013;3:139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
44.  Sharma A, Gokulchandran N, Sane H, Nagrajan A, Paranjape A, Kulkarni P, Shetty A, Mishra P, Kali M, Biju H. Autologous bone marrow mononuclear cell therapy for autism: an open label proof of concept study. Stem Cells Int. 2013;2013:623875.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 59]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
45.  Venugopal P, Balasubramanian S, Majumdar AS, Ta M. Isolation, characterization, and gene expression analysis of Wharton’s jelly-derived mesenchymal stem cells under xeno-free culture conditions. Stem Cells Cloning. 2011;4:39-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 16]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
46.  Cook MM, Futrega K, Osiecki M, Kabiri M, Kul B, Rice A, Atkinson K, Brooke G, Doran M. Micromarrows--three-dimensional coculture of hematopoietic stem cells and mesenchymal stromal cells. Tissue Eng Part C Methods. 2012;18:319-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 49]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
47.  Ghosh A, Michalon A, Lindemann L, Fontoura P, Santarelli L. Drug discovery for autism spectrum disorder: challenges and opportunities. Nat Rev Drug Discov. 2013;12:777-790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 80]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]