1
|
Schubart JR, Zare A, Fernandez-de-Castro RM, Figueroa HR, Sarel I, Tuchman K, Esposito K, Henderson FC, von Schwarz E. Safety and outcomes analysis: transcatheter implantation of autologous angiogenic cell precursors for the treatment of cardiomyopathy. Stem Cell Res Ther 2023; 14:308. [PMID: 37880753 PMCID: PMC10601268 DOI: 10.1186/s13287-023-03539-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Stem cell transplantation is an emerging therapy for severe cardiomyopathy, proffering stem cell recruitment, anti-apoptosis, and proangiogenic capabilities. Angiogenic cell precursors (ACP-01) are autologous, lineage-specific, cells derived from a multipotent progenitor cell population, with strong potential to effectively engraft, form blood vessels, and support tissue survival and regeneration. METHODS This IRB approved outcome analysis reports upon 74 consecutive patients who failed medical management for severe cardiomyopathy, and were selected to undergo transcatheter intramyocardial or intracoronary implantation of ACP-01. Serious adverse events (SAEs) were reported. Cell analysis was conducted for each treatment. The left ventricular ejection fraction (LVEF) was measured by multi-gated acquisition scan (MUGA) or echocardiogram at 4 months ± 1.9 months and 12 months ± 5.5 months. Patients reported quality of life statements at 6 months (± 5.6 months). RESULTS Fifty-four of 74 patients met requirements for inclusion (48 males and five females; age 68.1 ± 11.3 years). The mean treatment cell number of 57 × 106 ACP-01 included 7.7 × 106 CD34 + and 21 × 106 CD31 + cells with 97.6% viability. SAEs included one death (previously unrecognized silent MI), ventricular tachycardia (n = 2) requiring cardioversion, and respiratory infection (n = 2). LVEF in the ischemic subgroup (n = 41) improved by 4.7% ± 9.7 from pre-procedure to the first follow-up (4 months ± 1.9 months) (p < 0.004) and by 7.2% ± 10.9 at final follow-up (n = 25) at average 12 months (p < 0.004). The non-ischemic dilated cardiomyopathy subgroup (n = 8) improved by 7.5% ± 6.0 at the first follow-up (p < 0.017) and by 12.2% ± 6.4 at final follow-up (p < 0.003, n = 6). Overall improvement in LVEF from pre-procedure to post-procedure was significant (Fisher's exact test p < 0.004). LVEF improvement was most marked in the patients with the most severe cardiomyopathy (LVEF < 20%) improving from a mean 14.6% ± 3.4% pre-procedurally to 28.4% ± 8% at final follow-up. Quality of life statements reflected improvement in 33/50 (66%), no change in 14/50 (28%), and worse in 3/50 (6%). CONCLUSION Transcatheter implantation of ACP-01 for cardiomyopathy is safe and improves LVEF in the setting of ischemic and non-ischemic cardiomyopathy. The results warrant further investigation in a prospective, blinded, and controlled clinical study. TRIAL REGISTRATION IRB from Genetic Alliance #APC01-001, approval date July 25, 2022. Cardiomyopathy is common and associated with high mortality. Stem cell transplantation is an emerging therapy. Angiogenic cell precursors (ACP-01) are lineage-specific endothelial progenitors, with strong potential for migration, engraftment, angiogenesis, and support of tissue survival and regeneration. A retrospective outcomes analysis of 53 patients with ischemic and non-ischemic dilated cardiomyopathy undergoing transcatheter implantation of ACP-01 demonstrated improvements in the left ventricular ejection fraction of 7.2% ± 10.9 (p < 0.004) and 12.2% ± 6.4, respectively, at 12 months (± 5) follow-up. Quality of life statements reflected improvement in 33/50 (66%) patients.
Collapse
Affiliation(s)
- Jane R Schubart
- Penn State College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Amirhossein Zare
- Northern Ontario School of Medicine, Ontario, CA, USA
- Hemostemix Inc, Calgary, CA, Canada
| | | | | | | | - Kelly Tuchman
- The Metropolitan Neurosurgery Group, LLC, 1010 Wayne Ave Suite 420, Silver Spring, MD, 20910, USA.
| | - Kaitlyn Esposito
- The Bobby Jones Chiari Syringomyelia Foundation, New York, NY, USA
| | - Fraser C Henderson
- The Metropolitan Neurosurgery Group, LLC, 1010 Wayne Ave Suite 420, Silver Spring, MD, 20910, USA.
- Department Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA.
- Hemostemix Inc, Calgary, CA, Canada.
| | - Ernst von Schwarz
- School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Cedars Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
2
|
Bellon A, Feuillet V, Cortez-Resendiz A, Mouaffak F, Kong L, Hong LE, De Godoy L, Jay TM, Hosmalin A, Krebs MO. Dopamine-induced pruning in monocyte-derived-neuronal-like cells (MDNCs) from patients with schizophrenia. Mol Psychiatry 2022; 27:2787-2802. [PMID: 35365810 PMCID: PMC9156413 DOI: 10.1038/s41380-022-01514-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 02/05/2022] [Accepted: 02/25/2022] [Indexed: 01/10/2023]
Abstract
The long lapse between the presumptive origin of schizophrenia (SCZ) during early development and its diagnosis in late adolescence has hindered the study of crucial neurodevelopmental processes directly in living patients. Dopamine, a neurotransmitter consistently associated with the pathophysiology of SCZ, participates in several aspects of brain development including pruning of neuronal extensions. Excessive pruning is considered the cause of the most consistent finding in SCZ, namely decreased brain volume. It is therefore possible that patients with SCZ carry an increased susceptibility to dopamine's pruning effects and that this susceptibility would be more obvious in the early stages of neuronal development when dopamine pruning effects appear to be more prominent. Obtaining developing neurons from living patients is not feasible. Instead, we used Monocyte-Derived-Neuronal-like Cells (MDNCs) as these cells can be generated in only 20 days and deliver reproducible results. In this study, we expanded the number of individuals in whom we tested the reproducibility of MDNCs. We also deepened the characterization of MDNCs by comparing its neurostructure to that of human developing neurons. Moreover, we studied MDNCs from 12 controls and 13 patients with SCZ. Patients' cells differentiate more efficiently, extend longer secondary neurites and grow more primary neurites. In addition, MDNCs from medicated patients expresses less D1R and prune more primary neurites when exposed to dopamine. Haloperidol did not influence our results but the role of other antipsychotics was not examined and thus, needs to be considered as a confounder.
Collapse
Affiliation(s)
- Alfredo Bellon
- Department of Psychiatry and Behavioral Health, Penn State Hershey Medical Center, Hershey, PA, USA.
- Department of Pharmacology, Penn State Hershey Medical Center, Hershey, PA, USA.
| | - Vincent Feuillet
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014, Paris, France
| | - Alonso Cortez-Resendiz
- Department of Psychiatry and Behavioral Health, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Faycal Mouaffak
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
- Pôle de Psychiatrie d'Adultes 93G04, EPS Ville Evrard, Saint Denis, France
| | - Lan Kong
- Department of Public Health Sciences, Penn State Hershey Medical Center, Hershey, PA, USA
| | - L Elliot Hong
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Therese M Jay
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
| | - Anne Hosmalin
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014, Paris, France
| | - Marie-Odile Krebs
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
- Groupe-Hospitalo-Universitaire de Paris, Psychiatrie et Neuroscience, Pôle PEPIT, University of Paris, Paris, France
| |
Collapse
|
3
|
Zhao M, Zhang H, Zhen D, Huang M, Li W, Li Z, Liu Y, Xie Y, Zeng B, Wang Z, Huang B. Corneal Recovery Following Rabbit Peripheral Blood Mononuclear Cell-Amniotic Membrane Transplantation with Antivascular Endothelial Growth Factor in Limbal Stem Cell Deficiency Rabbits. Tissue Eng Part C Methods 2020; 26:541-552. [PMID: 33019886 DOI: 10.1089/ten.tec.2020.0209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Limbal stem cell deficiency (LSCD) is a refractory ocular surface disorder characterized by progressive corneal epithelial degeneration, conjunctivalization, and neovascularization, potentially leading to blindness. There are currently no effective therapeutic options for patients experiencing routine symptomatic treatment failure. Transplantation of amniotic membrane (AM) with adherent stem cells (but not bare AM transplantation alone) has shown promise in preclinical studies for ocular surface restoration. A major limitation, however, is finding a reliable stem cell source. Stem cells can be isolated from the peripheral blood mononuclear cell (PBMC) population, and these PBMC-derived stem cells have numerous advantages over allogeneic and other autologous stem cell types for therapeutic application, including relative ease of acquisition, nonimmunogenicity, and the absence of ethical issues associated with embryonic stem cells. Experiment: We examined the efficacy of autologous PBMC-AM sheet cultures combined with postoperative antiangiogenesis treatment for corneal restoration in LSCD model rabbits. Rabbit PBMCs (rPBMCs) were isolated, labeled with EdU for in vivo tracing, and then cultured on AMs in conditioned medium before transplantation. Rabbits were transplanted with bare AMs (group 1), rPBMC-AM sheets (group 2), or rPBMC-AM sheets plus postoperative treatment with the vascular endothelial growth factor antagonist bevacizumab (group 3). Corneal opacity and neovascularization were monitored by slit-lamp imaging for 8 weeks and corneas were examined histologically at 1 and 2 months. Results: Corneal opacity decreased in all three groups over 8 weeks, but was significantly lower in group 2 and even lower in group 3. Corneal neovascularization was significantly higher in group 1 throughout the observation period, and significantly lower in group 3 than group 1 and 2 by 8 weeks post-transplant. At 4 weeks, the corneal surface completed epithelialization (although thinner than normal) in group 3 but still patchy in groups 1 and 2. By 8 weeks, the epithelium in group 3 was complete and smooth, resembling a normal epithelium. Integrin β1 as a progenitor marker was also generally higher in groups 2 and 3. Conclusions: Autologous rPBMC-AM sheets with post-transplant topical bevacizumab can effectively facilitate corneal epithelium recovery in a LSCD model, suggesting clinical utility for LSCD-related ocular surface diseases. Impact statement Limbal stem cell deficiency (LSCD) increases corneal opacity and vascularization, resulting in severe visual impairment or even blindness. Traditional surgical limbal transplant is currently the main treatment option for LSCD, but carries the risks of rejection and immunosuppressant side effects. Autologous stem cell-based therapy is a promising alternative approach, but a reliable stem cell source is a major limitation. We report that transplantation of autologous rabbit peripheral blood mononuclear cell-amniotic membrane sheets plus antivascular endothelial growth factor restored avascular transparent cornea in a rabbit LSCD model. These results demonstrate a potentially effective approach for ocular surface reconstruction in bilateral LSCD.
Collapse
Affiliation(s)
- Minglei Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Hening Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Dongqin Zhen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | | | - Weihua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Zhiquan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Ying Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Yaojue Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Baozhu Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Zhichong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Bing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| |
Collapse
|
4
|
Bellon A, Wegener A, Lescallette AR, Valente M, Yang SK, Gardette R, Matricon J, Mouaffak F, Watts P, Vimeux L, Yun JK, Kawasawa YI, Clawson GA, Blandin E, Chaumette B, Jay TM, Krebs MO, Feuillet V, Hosmalin A. Transdifferentiation of Human Circulating Monocytes Into Neuronal-Like Cells in 20 Days and Without Reprograming. Front Mol Neurosci 2018; 11:323. [PMID: 30760979 PMCID: PMC6156467 DOI: 10.3389/fnmol.2018.00323] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/21/2018] [Indexed: 12/19/2022] Open
Abstract
Despite progress, our understanding of psychiatric and neurological illnesses remains poor, at least in part due to the inability to access neurons directly from patients. Currently, there are in vitro models available but significant work remains, including the search for a less invasive, inexpensive and rapid method to obtain neuronal-like cells with the capacity to deliver reproducible results. Here, we present a new protocol to transdifferentiate human circulating monocytes into neuronal-like cells in 20 days and without the need for viral insertion or reprograming. We have thoroughly characterized these monocyte-derived-neuronal-like cells (MDNCs) through various approaches including immunofluorescence (IF), flow cytometry, qRT-PCR, single cell mRNA sequencing, electrophysiology and pharmacological techniques. These MDNCs resembled human neurons early in development, expressed a variety of neuroprogenitor and neuronal genes as well as several neuroprogenitor and neuronal proteins and also presented electrical activity. In addition, when these neuronal-like cells were exposed to either dopamine or colchicine, they responded similarly to neurons by retracting their neuronal arborizations. More importantly, MDNCs exhibited reproducible differentiation rates, arborizations and expression of dopamine 1 receptors (DR1) on separate sequential samples from the same individual. Differentiation efficiency measured by cell morphology was on average 11.9 ± 1.4% (mean, SEM, n = 38,819 cells from 15 donors). To provide context and help researchers decide which in vitro model of neuronal development is best suited to address their scientific question,we compared our results with those of other in vitro models currently available and exposed advantages and disadvantages of each paradigm.
Collapse
Affiliation(s)
- Alfredo Bellon
- Penn State Hershey Medical Center, Department of Psychiatry, Hershey, PA, United States.,Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA, United States.,INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France.,Centre Hospitalier Sainte-Anne, Faculté de Médecine Paris Descartes, Service Hospitalo-Universitaire-S14, Paris, France
| | - Amelie Wegener
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France
| | - Adam R Lescallette
- Penn State Hershey Medical Center, Department of Psychiatry, Hershey, PA, United States
| | - Michael Valente
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France
| | - Seung-Kwon Yang
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France
| | - Robert Gardette
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France
| | - Julien Matricon
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France
| | - Faycal Mouaffak
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France.,Centre Hospitalier Sainte-Anne, Faculté de Médecine Paris Descartes, Service Hospitalo-Universitaire-S14, Paris, France
| | - Paula Watts
- Sky Ridge Medical Center, Department of Internal Medicine, Lone Tree, CO, United States
| | - Lene Vimeux
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France
| | - Jong K Yun
- Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA, United States
| | - Yuka Imamura Kawasawa
- Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA, United States.,Penn State Hershey Medical Center, Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, Hershey, PA, United States
| | - Gary A Clawson
- Gittlen Cancer Research Laboratories, Department of Pathology, Penn State University College of Medicine, Hershey, PA, United States
| | - Elisabeta Blandin
- Penn State Hershey Medical Center, Department of Psychiatry, Hershey, PA, United States.,Penn State Hershey Medical Center, Neural & Behavioral Sciences, Hershey, PA, United States
| | - Boris Chaumette
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France.,Centre Hospitalier Sainte-Anne, Faculté de Médecine Paris Descartes, Service Hospitalo-Universitaire-S14, Paris, France.,Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Therese M Jay
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France
| | - Marie-Odile Krebs
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France.,Centre Hospitalier Sainte-Anne, Faculté de Médecine Paris Descartes, Service Hospitalo-Universitaire-S14, Paris, France
| | - Vincent Feuillet
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France
| | - Anne Hosmalin
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France
| |
Collapse
|
5
|
Tara S, Krishnan LK. Differentiation of circulating neural progenitor cells in vitro on fibrin-based composite -matrix involves Wnt- β-catenin-like signaling. J Cell Commun Signal 2018; 13:27-38. [PMID: 29856041 DOI: 10.1007/s12079-018-0467-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 04/19/2018] [Indexed: 02/07/2023] Open
Abstract
Isolation of progenitors with regenerative potential and their in vitro induction to specific lineage may be necessary for effective cell transplantation outcome. Earlier, we standardized specific niche for derivation of neural progenitor cells (NPCs) from circulating mononuclear cells to neural like cells (NLC) in vitro, for applications in neural regeneration. The current study analysed the prospective involvement of signaling mechanism for in vitro lineage commitment of circulating NPCs. Preferred mechanism selected was Wnt-like signaling because this is one of the pathways implicated in the central nervous system (CNS) development and homeostasis. We sought to determine the activation of Wnt3a-specific genes in the standardized NPC culture system. To start with, it was found that when standardized NPC culture niche was supplemented with Wnt 3a protein, no additional morphological changes happen. Chemical inhibitors of the pathway retarded NPC to NLC conversion both in the absence and presence of supplemented Wnt-3a. In earlier studies, involvement of the niche constituents- fibronectin (FN), laminin (La) and fibrin (Fib)- for NPC growth and differentiation was established. In an attempt to study the role of these adhesive proteins by adding antibodies against FN, La & Fib together, molecular level signaling changes seen were comparable to that occur in response to Wnt3a chemical inhibitor. Therefore, induction of Wnt 3a-like signal from the matrix-dependent niche constituents may be implicated in the differentiation of NPC to NLC. The results substantiate the potential applications of the fibrin-based composite niche in neural engineering for regeneration.
Collapse
Affiliation(s)
- S Tara
- Division of Thrombosis Research, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Trivandrum, 695012, India
| | - Lissy K Krishnan
- Division of Thrombosis Research, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Trivandrum, 695012, India.
| |
Collapse
|
6
|
Eve DJ, Sanberg PR, Buzanska L, Sarnowska A, Domanska-Janik K. Human Somatic Stem Cell Neural Differentiation Potential. Results Probl Cell Differ 2018; 66:21-87. [DOI: 10.1007/978-3-319-93485-3_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
7
|
Tara S, Krishnan LK. Bioengineered fibrin-based niche to direct outgrowth of circulating progenitors into neuron-like cells for potential use in cellular therapy. J Neural Eng 2015; 12:036011. [PMID: 25946462 DOI: 10.1088/1741-2560/12/3/036011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Autologous cells are considered to be the best choice for use in transplantation therapy. However, the challenges and risks associated with the harvest of transplantable autologous cells limit their successful therapeutic application. The current study explores the possibility of isolating neural progenitor cells from circulating multipotent adult progenitor cells for potential use in cell-based and patient-specific therapy for neurological diseases. APPROACH To enable the selection of neural progenitor cells from human peripheral blood mononuclear cells, and to support their lineage maintenance, the composition of a fibrin-based niche was optimized. Morphological examination and specific marker analysis were carried out, employing a qualitative/quantitative polymerase chain reaction followed by immunocytochemistry to: (i) characterize neural progenitor cells in culture; (ii) monitor proliferation/survival; and (iii) track their differentiation status. MAIN RESULTS The presence of neural progenitors in circulation was confirmed by the presence of nestin(+) cells at the commencement of the culture. The isolation, proliferation and differentiation of circulating neural progenitors to neuron-like cells were directed by the engineered niche. Neural cell isolation to near homogeneity was confirmed by the expression of β-III tubulin in ∼95% of cells, whereas microtubule associated protein-2 expression confirmed their ability to differentiate. The concentration of potassium chloride in the niche was found to favour neuron-like cell lengthening, cell-cell contact, and expressions of synaptophysin and tyrosine hydroxylase. SIGNIFICANCE The purpose of this research was to find out if peripheral blood could serve as a potential source of neural progenitors for cell based therapy. The study established that neural progenitors could be selectively isolated from peripheral blood mononuclear cells using a biomimetic niche. The selected cells could multiply and slowly differentiate into neuron-like cells. These neuron-like cells expressed functional proteins-tyrosine hydroxylase and synaptophysin. Early progenitors that proliferate while expressing β-III tubulin could be harvested from the culture, suggesting their potential use in cell transplantation therapy.
Collapse
Affiliation(s)
- S Tara
- Thrombosis Research Unit, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India 695012
| | | |
Collapse
|
8
|
Porat Y, Abraham E, Karnieli O, Nahum S, Woda J, Zylberberg C. Critical elements in the development of cell therapy potency assays for ischemic conditions. Cytotherapy 2015; 17:817-31. [PMID: 25728414 DOI: 10.1016/j.jcyt.2014.08.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/22/2014] [Accepted: 08/26/2014] [Indexed: 12/28/2022]
Abstract
A successful potency assay for a cell therapy product (CTP) used in the treatment of ischemic conditions should quantitatively measure relevant biological properties that predict therapeutic activity. This is especially challenging because of numerous degrees of complexity stemming from factors that include a multifactorial complex mechanism of action, cell source, inherent cell characteristics, culture method, administration mode and the in vivo conditions to which the cells are exposed. The expected biological function of a CTP encompasses complex interactions that range from a biochemical, metabolic or immunological activity to structural replacement of damaged tissue or organ. Therefore, the requirements for full characterization of the active substance with respect to biological function could be taxing. Moreover, the specific mechanism of action is often difficult to pinpoint to a specific molecular entity; rather, it is more dependent on the functionality of the cellular components acting in a in a multifactorial fashion. In the case of ischemic conditions, the cell therapy mechanism of action can vary from angiogenesis, vasculogenesis and arteriogenesis that may activate different pathways and clinical outcomes. The CTP cellular attributes with relation to the suggested mechanism of action can be used for the development of quantitative and reproducible analytical potency assays. CTPs selected and released on the basis of such potency assays should have the highest probability of providing meaningful clinical benefit for patients. This White Paper will discuss and give examples for key elements in the development of a potency assay for treatment of ischemic disorders treated by the use of CTPs.
Collapse
Affiliation(s)
- Yael Porat
- BioGenCell Ltd, Hematology BGC Stem Cell Research, Sanz Medical Center Laniado Hospital, Netanya, Israel
| | | | | | | | | | | |
Collapse
|
9
|
Porat Y, Assa-Kunik E, Belkin M, Krakovsky M, Lamensdorf I, Duvdevani R, Sivak G, Niven MJ, Bulvik S. A novel potential therapy for vascular diseases: blood-derived stem/progenitor cells specifically activated by dendritic cells. Diabetes Metab Res Rev 2014; 30:623-34. [PMID: 24638886 DOI: 10.1002/dmrr.2543] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 02/26/2014] [Accepted: 03/03/2014] [Indexed: 01/01/2023]
Abstract
BACKGROUND Vascular diseases are a major cause of morbidity and mortality, particularly in diabetic patients. Stem/progenitor cell treatments with bone marrow-derived cells show safety and promising outcomes, albeit not without some preprocedural adverse events related to cell collection and mobilization. We describe a novel technology for generating a therapeutic population (BGC101) of enriched endothelial progenitor cells (EPCs) from non-mobilized blood, using dendritic cells to specifically direct stem/progenitor cell activity in vitro. METHODS AND RESULTS Selected immature plasmacytoid and myeloid dendritic cells from 24 healthy and two diabetic donors were activated with anti-inflammatory and pro-angiogenic molecules to induce specific activation signals. Co-culturing of activated dendritic cells with stem/progenitor cells for 12-66 h generated 83.7 ± 7.4 × 10(6) BGC101 cells with 97% viability from 250 mL of blood. BGC101, comprising 52.4 ± 2.5% EPCs (expressing Ulex-lectin, AcLDL uptake, Tie2, vascular endothelial growth factor receptor 1 and 2, and CD31), 16.1 ± 1.9% stem/progenitor cells (expressing CD34 and CD184) and residual B and T helper cells, demonstrated angiogenic and stemness potential and secretion of interleukin-8, interleukin-10, vascular endothelial growth factor and osteopontin. When administered to immunodeficient mice with limb ischemia (n = 40), BGC101 yielded a high safety profile and significantly increased blood perfusion, capillary density and leg function after 21 days. Cell tracking and biodistribution showed that engraftment was restricted to the ischemic leg. CONCLUSIONS These observations provide preliminary evidence that alternatively activated dendritic cells can promote the generation of EPC-enriched stem/progenitor cells within a 1-day culture. The resulting product BGC101 has the potential for treatment of various vascular conditions such as coronary heart disease, stroke and peripheral ischemia.
Collapse
Affiliation(s)
- Yael Porat
- BioGenCell Ltd, Sanz Medical Center, Laniado Hospital, Netanya, Israel; Sanz Medical Center, Laniado Hospital, Netanya, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Li H, Li J, Sheng W, Sun J, Ma X, Chen X, Bi J, Zhao Y, Li X. Astrocyte-like cells differentiated from a novel population of CD45-positive cells in adult human peripheral blood. Cell Biol Int 2014; 39:84-93. [PMID: 25077697 PMCID: PMC4410680 DOI: 10.1002/cbin.10355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 06/11/2014] [Indexed: 01/06/2023]
Abstract
We have previously reported a novel CD45-positive cell population called peripheral blood insulin-producing cells (PB-IPCs) and its unique potential for releasing insulin in vitro. Despite the CD45-positive phenotype and self-renewal ability, PB-IPCs are distinguished from hemopoietic and endothelial progenitor cells (EPCs) by some characteristics, such as a CD34-negative phenotype and different culture conditions. We have further identified the gene profiles of the embryonic and neural stem cells, and these profiles include Sox2, Nanog, c-Myc, Klf4, Notch1 and Mash1. After treatment with all-trans retinoic acid (ATRA) in vitro, most PB-IPCs exhibited morphological changes that included the development of elongated and branched cell processes. In the process of induction, the mRNA expression of Hes1 was robustly upregulated, and a majority of cells acquired some astrocyte-associated specific phenotypes including anti-glial fibrillary acidic protein (GFAP), CD44, Glutamate-aspartate transporter (GLAST) and S100β. In spite of the deficiency of glutamate uptaking, the differentiated cells significantly relaxed the regulation of the expression of brain-derived neurotrophic factor (BDNF) mRNA. This finding demonstrates that PB-IPCs could be induced into a population of astrocyte-like cells and enhanced the neurotrophic potential when the state of proliferation was limited by ATRA, which implies that this unique CD45+ cell pool may have a protective role in some degenerative diseases of the central nervous system (CNS).
Collapse
Affiliation(s)
- Heng Li
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, China
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Zhang Y, Luo Y, Li K, Zhang M, Huang B, Peng Y, Wang W, Li W, Liu Y. Pre-induced adult human peripheral blood mononuclear cells migrate widely into the degenerative retinas of rd1 mice. Cytotherapy 2014; 15:1416-25. [PMID: 24094491 DOI: 10.1016/j.jcyt.2013.05.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 05/11/2013] [Accepted: 05/22/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND AIMS Recent advances in stem cell research have raised the possibility of stem cells repairing or replacing retinal photoreceptor cells that are either dysfunctional or lost in many retinal diseases. Various types of stem cells have been used to replace retinal photoreceptor cells. Recently, peripheral blood stem cells, a small proportion of pluripotent stem cells, have been reported to mainly exist in the peripheral blood mononuclear cells (PBMCs). METHODS In this study, the effects of pre-induced adult human PBMCs (hPBMCs) on the degenerative retinas of rd1 mice were investigated. Freshly isolated adult hPBMCs were pre-induced with the use of the conditioned medium of rat retinas for 4 days and were then labeled with chloromethyl-benzamidodialkylcarbocyanine (CM-DiI) and then transplanted into the subretinal space of the right eye of rd1 mice through a trans-scleral approach. The right eyes were collected 30 days after transplantation. The survival and migration of the transplanted cells in host retinas were investigated by whole-mount retinas, retinal frozen sections and immunofluorescent staining. RESULTS After subretinal transplantation, pre-induced hPBMCs were able to survive and widely migrate into the retinas of rd1 mice. A few CM-DiI-labeled cells migrated into the inner nuclear layer and the retinal ganglion cell layer. Some transplanted cells in the subretinal space of rd1 host mice expressed the human photoreceptor-specific marker rhodopsin. CONCLUSIONS This study suggests that pre-induced hPBMCs may be a potential cell source of cell replacement therapy for retinal degenerative diseases.
Collapse
Affiliation(s)
- Yichi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Peripheral blood-derived autologous stem cell therapy for the treatment of patients with late-stage peripheral artery disease-results of the short- and long-term follow-up. Cytotherapy 2014; 15:1245-52. [PMID: 23993298 DOI: 10.1016/j.jcyt.2013.05.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 05/13/2013] [Accepted: 05/22/2013] [Indexed: 11/20/2022]
Abstract
BACKGROUND AIMS Regeneration of the occluded peripheral arteries by autologous stem cell therapy is an emerging treatment modality for no-option patients with peripheral artery disease (PAD). The purpose of this study was to assess safety and efficacy of in vitro-expanded, peripheral blood-derived, autologous stem cells (VesCell) in no-option patients with PAD. METHODS A phase II, open-label, randomized clinical study was performed on 20 patients to investigate the safety and efficacy of VesCell therapy at 1 and 3 months of follow-up. The long-term (2 years) efficacy of the therapy was also evaluated. RESULTS No side effects of VesCell therapy were found. During the 3 month follow-up in the control group, one death occurred and six major amputations were performed; in the treated group, there were no deaths or major amputations. The difference of limb loss is significant between the two groups. At 2-year follow-up in the control group, two deaths and six major amputations occurred; in the treated group, there were three major amputations. At 3-month follow-up, the change in hemodynamic parameters showed a significant increase in the treated group over the control group; in the treated group, further improvement was detected at 2 years. As the result of the VesCell treatment, change in pain score, wound healing and walking ability test showed an improvement compared with the control group; at 2 years, incremental improvement was observed. CONCLUSIONS Peripheral blood-derived, in vitro-expanded autologous angiogenic precursor therapy appears to be a safe, promising and effective adjuvant therapy for PAD patients.
Collapse
|
13
|
Human angiogenic cell precursors restore function in the infarcted rat heart: A comparison of cell delivery routes. Eur J Heart Fail 2014; 10:525-33. [DOI: 10.1016/j.ejheart.2008.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Revised: 03/10/2008] [Accepted: 04/09/2008] [Indexed: 11/17/2022] Open
|
14
|
Derivation of neural stem cells from human adult peripheral CD34+ cells for an autologous model of neuroinflammation. PLoS One 2013; 8:e81720. [PMID: 24303066 PMCID: PMC3841177 DOI: 10.1371/journal.pone.0081720] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 10/23/2013] [Indexed: 12/31/2022] Open
Abstract
Proinflammatory factors from activated T cells inhibit neurogenesis in adult animal brain and cultured human fetal neural stem cells (NSC). However, the role of inhibition of neurogenesis in human neuroinflammatory diseases is still uncertain because of the difficulty in obtaining adult NSC from patients. Recent developments in cell reprogramming suggest that NSC may be derived directly from adult fibroblasts. We generated NSC from adult human peripheral CD34+ cells by transfecting the cells with Sendai virus constructs containing Sox2, Oct3/4, c-Myc and Klf4. The derived NSC could be differentiated to glial cells and action potential firing neurons. Co-culturing NSC with activated autologous T cells or treatment with recombinant granzyme B caused inhibition of neurogenesis as indicated by decreased NSC proliferation and neuronal differentiation. Thus, we have established a unique autologous in vitro model to study the pathophysiology of neuroinflammatory diseases that has potential for usage in personalized medicine.
Collapse
|
15
|
Ben Aharon I, Bar Joseph H, Tzabari M, Shenkman B, Farzam N, Levi M, Shalgi R, Stemmer SM, Savion N. Doxorubicin-induced vascular toxicity--targeting potential pathways may reduce procoagulant activity. PLoS One 2013; 8:e75157. [PMID: 24073244 PMCID: PMC3779248 DOI: 10.1371/journal.pone.0075157] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 08/12/2013] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Previous study in mice using real-time intravital imaging revealed an acute deleterious effect of doxorubicin (DXR) on the gonadal vasculature, as a prototype of an end-organ, manifested by a reduction in blood flow and disintegration of the vessel wall. We hypothesized that this pattern may represent the formation of microthrombi. We aimed to further characterize the effect of DXR on platelets' activity and interaction with endothelial cells (EC) and to examine potential protectants to reduce DXR acute effect on the blood flow. METHODS The effect of DXR on platelet adhesion and aggregation were studied in vitro. For in vivo studies, mice were injected with either low molecular weight heparin (LMWH; Enoxaparin) or with eptifibatide (Integrilin(©)) prior to DXR treatment. Testicular arterial blood flow was examined in real-time by pulse wave Doppler ultrasound. RESULTS Platelet treatment with DXR did not affect platelet adhesion to a thrombogenic surface but significantly decreased ADP-induced platelet aggregation by up to 40% (p<0.001). However, there was a significant increase in GPIIbIIIa-mediated platelet adhesion to DXR-exposed endothelial cells (EC; 5.7-fold; p<0.001) reflecting the toxic effect of DXR on EC. The testicular arterial blood flow was preserved in mice pre-treated with LMWH or eptifibatide prior to DXR (P<0.01). CONCLUSIONS DXR-induced acute vascular toxicity may involve increased platelet-EC adhesion leading to EC-bound microthrombi formation resulting in compromised blood flow. Anti-platelet/anti-coagulant agents are effective in reducing the detrimental effect of DXR on the vasculature and thus may serve as potential protectants to lessen this critical toxicity.
Collapse
Affiliation(s)
- Irit Ben Aharon
- Institute of Oncology, Davidoff Center and Rabin Medical Center, Petah-Tiqva and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| | - Hadas Bar Joseph
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moran Tzabari
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Boris Shenkman
- Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel Hashomer, Israel
| | - Nahid Farzam
- Goldschleger Eye Research Institute and Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mattan Levi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ruth Shalgi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Salomon M. Stemmer
- Institute of Oncology, Davidoff Center and Rabin Medical Center, Petah-Tiqva and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Naphtali Savion
- Goldschleger Eye Research Institute and Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
16
|
Eve DJ, Marty PJ, McDermott RJ, Klasko SK, Sanberg PR. Stem Cell Research and Health Education. AMERICAN JOURNAL OF HEALTH EDUCATION 2013. [DOI: 10.1080/19325037.2008.10599033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- David J. Eve
- a Center of Excellence for Aging and Brain Repair , University of South Florida College of Medicin, Department of Neurosurgery , 12901 Bruce B. Downs Blvd. (MDC 078), Tampa , FL , 33612
| | - Phillip J. Marty
- b Department of Community and Family Health , University of South Florida, College of Public Health
| | - Robert J. McDermott
- b Department of Community and Family Health , University of South Florida, College of Public Health
| | | | - Paul R. Sanberg
- d Center of Excellence for Aging and Brain Repair , University of South Florida
| |
Collapse
|
17
|
Peripheral blood stem cells: phenotypic diversity and potential clinical applications. Stem Cell Rev Rep 2012; 8:917-25. [PMID: 22451417 DOI: 10.1007/s12015-012-9361-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A small proportion of cells in peripheral blood are actually pluripotent stem cells. These peripheral blood stem cells (PBSCs) are thought to be heterogeneous and could be exploited for a variety of clinical applications. The exact number of distinct populations is unknown. It is likely that individual PBSC populations detected by different experimental strategies are similar or overlapping but have been assigned different names. In this mini review, we divide PBSCs into seven groups: hematopoietic stem cells (HSCs), CD34- stem cells, CD14+ stem cells, mesenchymal stem cells (MSCs), very small embryonic-like (VSEL) stem cells, endothelial progenitor cells (EPCs), and other pluripotent stem cells. We review the major characteristics of these stem/progenitor cell populations and their potential applications in ophthalmology.
Collapse
|
18
|
Shirvan MK, Alamdari DH, Mahboub MD, Ghanadi A, Rahimi HR, Seifalian AM. A novel cell therapy for stress urinary incontinence, short-term outcome. Neurourol Urodyn 2012; 32:377-82. [PMID: 22972395 DOI: 10.1002/nau.22301] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 07/30/2012] [Indexed: 12/27/2022]
Abstract
AIMS The aim of this study was the safety assessment of urethra injections of autologous total nucleated cells (TNCs) along with platelets, which focused on the outcome over a 6 month period. METHODS An open, prospective study was conducted on 9 patients with severe stress urinary incontinence (SUI). At the baseline, 1, 3, and 6 months after external urethral sphincteric and submucosal injections of autologous TNCs along with platelets, the patients were assessed according to cough tests, Q-Tip tests, urodynamics, 1 hr pad tests, upper tract ultrasonography (UTU), post voiding residue (PVR), International Consultation on Incontinence Questionnaire-Urinary incontinence (ICIQ-UI), and International Consultation on Incontinence Modular Questionnaire-Quality of Life (ICIQ-QOL). On the 3rd month post-injection, the maximum urethral closure pressure (MUCP) and abdominal leak point pressure (ALPP) were measured in one patient with intrinsic sphincteric deficiency (ISD; the baseline: ALPP < 60 and MUCP < 30 cmH(2)O). RESULTS No complications were observed after injection. At 6-months' follow up (F/U), all the patients considered themselves clinically cured with eight women completely continent and one marked improvement. Mean age was 48.9 ± 13.8 years. Before the injection, urodynamics, UTU, and PVR were normal and cough tests, 1 hr pad tests were positive in patients. At 1, 3, and 6 months post-injection, there was a significant improvement in ICIQ-UI, ICIQ-QOL (P < 0.05). UTU and PVR were normal, cough tests, and 1 hr pad tests were negative, except for ISD patient with severe coughs (at month 3: ALPP = 92 and MUCP > 30 cmH(2)O). CONCLUSION Cell therapy consisting of intrasphincteric and submucosal injections of autologous TNCs along with platelets in SUI patients is a feasible and safe procedure. The results point out those subjects cured or with marked improvement after 6 months F/U.
Collapse
Affiliation(s)
- Maliheh Keshvari Shirvan
- Department of Urology, Imam Reza Academic Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | | | | | | |
Collapse
|
19
|
Czeiger D, Dukhno O, Douvdevani A, Porat Y, Shimoni D, Fulga V, Ament JD, Shaked G. Transient extremity ischemia augments CD34+ progenitor cell availability. Stem Cell Rev Rep 2011; 7:639-45. [PMID: 21318598 DOI: 10.1007/s12015-011-9234-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Peripheral blood is an easily accessed source for stem cell production; however, the number of cells produced is relatively low. We hypothesized that ischemic preconditioning may serve as a safe method to increase the number of CD34+ cells that can be harvested and cultured in a short period. This study was conducted to test this hypothesis by examining the safety and efficacy of brief, transient ischemia of the lower limbs to augment the number of cells that can be produced from blood of healthy volunteers. Following induction of ischemia, blood samples were withdrawn at baseline, 30 min, 12 h and 24 h. The number of progenitor cells was determined by flow cytometry after the harvested cells were cultured for 5 days. We also analyzed the blood samples to determine IL-8 and VEGF concentrations. No serious adverse events were observed. The total number of cells increased from 0.46 ± 0.1 × 10(6) cells/ml in the pretreatment blood samples to 0.7 ± 0.1 × 10(6) cells/ml in blood taken 12 h after the conclusion of transient ischemia, p = 0.0029. The number of CD34+ cells increased from 4.23 ± 0.8 × 10(4) cells/ml in the pretreatment samples to 7.17 ± 1.34 × 10(4) cells/ml in blood taken 12 h after ischemia, p = 0.0001. The harvested stem cells maintained their ability to construct tubular structures. The augmentation in the number of CD34+ cells was positively correlated with the increase of IL-8, but not with VEGF concentrations. Ischemic preconditioning is a safe and effective technique to increase the availability of stem cells for therapeutic purposes.
Collapse
Affiliation(s)
- David Czeiger
- Soroka University Medical Center, Beer Sheva, Israel
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
The importance and essential functions of glial cells in the nervous system are now beginning to be understood and appreciated. Glial cell lines have been instrumental in the elucidation of many of these properties. In this Overview, the origin and properties of most of the existing cell lines for the major glial types: oligodendroglia, astroglia, microglia and Schwann cells, are documented. Particular emphasis is given to the culture conditions for each cell line and the degree to which the line can differentiate in vitro and in vivo. The major molecular markers for each glial cell lines are indicated. Finally, methods by which the glial cell lines have been developed are noted and the future directions of glial cell line research are discussed.
Collapse
|
21
|
Jose A, Krishnan LK. Effect of matrix composition on differentiation of nestin-positive neural progenitors from circulation into neurons. J Neural Eng 2010; 7:036009. [PMID: 20479522 DOI: 10.1088/1741-2560/7/3/036009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The human peripheral blood mononuclear cell has a mixture of progenitor cells with potential to differentiate into a wide range of lineages. The ability of hematopoietic tissue-derived adult stem cells to differentiate into neural progenitor cells offers an alternative to embryonic stem cells as a viable source for cell transplantation therapies to cure neurodegenerative diseases. This approach could lead to the use of autologous progenitors from blood circulation; however, due to the limited numbers available, in vitro cell expansion may be indispensable. In addition, for successful transplantation there is the requirement of a delivery matrix on which cells can survive and differentiate. In this context we carried out this study to identify a suitable biodegradable matrix on which progenitor cells can home, multiply and differentiate. We designed different compositions of the biomimetic matrix containing fibrin, fibronectin, gelatin, growth factors, laminin and hyaluronic acid. The attached cells expressed proliferation markers in initial periods of culture and between days 6 and 9 in culture they differentiated into neurons and/or astrocytes. The differentiation of progenitors into neurons and asterocyte on the composed matrix was established by morphological and immunochemical analysis. Flow cytometric analysis of cells in culture was employed to track development of neurons which expressed an early marker beta-tubulin3 and a terminal marker microtubule-associated protein-2 at a later culture period. In vitro experiments indicate that a highly specific niche consisting of various components of the extracellular matrix, including hyaluronic acid, promote cell homing, survival and differentiation.
Collapse
Affiliation(s)
- Anumol Jose
- Thrombosis Research Unit, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Biomedical Technology Wing, Thiruvananthapuram-12, Kerala, India
| | | |
Collapse
|
22
|
Tarella C, Rutella S, Gualandi F, Melazzini M, Scimè R, Petrini M, Moglia C, Ulla M, Omedé P, Bella VL, Corbo M, Silani V, Siciliano G, Mora G, Caponnetto C, Sabatelli M, Chiò A. Consistent bone marrow-derived cell mobilization following repeated short courses of granulocyte–colony-stimulating factor in patients with amyotrophic lateral sclerosis: results from a multicenter prospective trial. Cytotherapy 2010; 12:50-9. [DOI: 10.3109/14653240903300682] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
23
|
Arom KV, Ruengsakulrach P, Belkin M, Tiensuwan M. Intramyocardial angiogenic cell precursors in nonischemic dilated cardiomyopathy. Asian Cardiovasc Thorac Ann 2009; 17:382-8. [PMID: 19713335 DOI: 10.1177/0218492309338105] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To determine the efficacy of intramyocardial injection of angiogenic cell precursors in nonischemic dilated cardiomyopathy, 35 patients with nonischemic dilated cardiomyopathy underwent injections of angiogenic cell precursors into the left ventricle (cell group). Seventeen patients with nonischemic dilated cardiomyopathy were matched from the heart failure database to form a control group that was treated medically. Angiogenic cell precursors were obtained from autologous blood, cultured in vitro, and injected into all free-wall areas of the left ventricle in the cell group. After these injections, New York Heart Association functional class improved significantly by 1.1 +/- 0.7 classes at 284.7 +/- 136.2 days, and left ventricular ejection fraction improved in 71.4% of patients (25/35); the mean increase in left ventricular ejection fraction was 4.4% +/- 10.6% at 192.7 +/- 135.1 days. Improved quality of life was demonstrated by better physical function, role-physical, general health, and vitality domains in a short-form health survey at the 3-month follow-up. In the control group, there were no significant improvements in left ventricular ejection fraction or New York Heart Association class which increased by 0.6 +/- 0.8 classes. It was concluded that intramyocardial angiogenic cell precursor injection is probably effective in the treatment of nonischemic dilated cardiomyopathy.
Collapse
Affiliation(s)
- Kitipan V Arom
- Cardiovascular Surgery, Bangkok Heart Hospital, Bangkok, Thailand.
| | | | | | | |
Collapse
|
24
|
Stem cell therapy for end-stage heart failure: indispensable role for the cell? Curr Opin Organ Transplant 2009; 14:560-5. [DOI: 10.1097/mot.0b013e328330389e] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
25
|
Devanesan A, Laughlan K, Girn H, Homer-Vanniasinkam S. Endothelial Progenitor Cells as a Therapeutic Option in Peripheral Arterial Disease. Eur J Vasc Endovasc Surg 2009; 38:475-81. [DOI: 10.1016/j.ejvs.2009.05.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2009] [Accepted: 05/29/2009] [Indexed: 10/20/2022]
|
26
|
Arom KV, Ruengsakulrach P, Chaothawee L. Improvement in the Severity of Mitral Regurgitation after Direct Intramyocardial Injection of Angiogenic Cell Precursor. INNOVATIONS-TECHNOLOGY AND TECHNIQUES IN CARDIOTHORACIC AND VASCULAR SURGERY 2009. [DOI: 10.1177/155698450900400511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
27
|
Improvement in the severity of mitral regurgitation after direct intramyocardial injection of angiogenic cell precursor. INNOVATIONS-TECHNOLOGY AND TECHNIQUES IN CARDIOTHORACIC AND VASCULAR SURGERY 2009; 4:291-3. [PMID: 22437170 DOI: 10.1097/imi.0b013e3181bbe750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We reported a case of dilated cardiomyopathy and moderate-severe mitral regurgitation (MR) who we treated by surgical direct intramyocardial angiogenic cell precursors injection. The patient was a New York Heart Association functional class III-IV, 56 year old man, who presented with end-stage congestive heart failure, moderate/severe mitral regurgitation, and myocardial fibrosis with the left ventricular ejection fraction of 13%. After he underwent direct surgical intramyocardial cell implantation, the myocardial fibrosis was resolved at 3 months follow-up. The severity of MR reduced to moderate and mild at 3 and 9 months, respectively. The left ventricular function gradually improved up to 53% at 19 months. To our knowledge, this is one of the only reports of successful direct surgical intramyocardial peripheral blood stem cell implantation to treat MR in dilated cardiomyopathy patient.
Collapse
|
28
|
Kiatpongsan S, Sipp D. MEDICINE: Monitoring and Regulating Offshore Stem Cell Clinics. Science 2009; 323:1564-5. [DOI: 10.1126/science.1168451] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
29
|
Arom KV, Ruengsakulrach P, Jotisakulratana V. Intramyocardial angiogenic cell precursor injection for cardiomyopathy. Asian Cardiovasc Thorac Ann 2008; 16:143-8. [PMID: 18381874 DOI: 10.1177/021849230801600213] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Stem cell therapy for heart failure is a rapidly progressing field. The objective of this study was to assess the safety, and short-term results of thoracoscopic direct injection of angiogenic cell precursors into patients with endstage cardiomyopathy. Cells were obtained from the patient's own blood, avoiding immunological concerns. The number of cells prior to injection was 29.1 +/- 18.9 x10(6). Forty-one patients with cardiomyopathy (mean age, 58.5 +/- 14.3 years) underwent stem cell injection; 21 had dilated cardiomyopathy and 20 had ischemic cardiomyopathy. Overall ejection fraction improved significantly by 4.8% +/- 7.5% at 149 +/- 98 days postoperatively. It increased from 25.9% +/- 8.6% to 28.7% +/- 9.8% in dilated cardiomyopathy, and from 26.6% +/- 5.8% to 33.6% +/- 7.8% in ischemic cardiomyopathy. New York Heart Association functional class was significantly better at 2 months in both groups. It was concluded that thoracoscopic intramyocardial angiogenic cell precursor injection is feasible and safe in patients with cardiomyopathy. The early results are good, and phase II trials are in progress.
Collapse
Affiliation(s)
- Kitipan V Arom
- Bangkok Heart Hospital, 2 Soi Soonvijai 7, New Petchburi Road, Bangkok 10320, Thailand.
| | | | | |
Collapse
|
30
|
Ballard VLT, Edelberg JM. Stem cells for cardiovascular repair - the challenges of the aging heart. J Mol Cell Cardiol 2008; 45:582-92. [PMID: 18396293 DOI: 10.1016/j.yjmcc.2008.02.277] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 02/22/2008] [Accepted: 02/22/2008] [Indexed: 01/04/2023]
Abstract
The discovery of extracardiac progenitor cells and resident cardiac stem cells in recent years has led to a great deal of interest in the development of therapeutic strategies that target these endogenous cell sources for promotion of cardiovascular repair mechanisms in the diseased heart. Cardiovascular risk increases with age and among many factors, the age-associated decline in cardiac and vascular regenerative capacity may contribute to the progressive deterioration of cardiovascular health. Thus, understanding the mechanisms which underlie the dysregulation of cardiac stem and progenitor cells may lead to the identification of novel targets and approaches to reverse this decline. In this review, we outline the current knowledge about cardiac stem and progenitor cells, their contribution to cardiovascular regenerative processes and factors that may affect their decreased function in aging individuals. Moreover, we describe the therapeutic strategies that are currently being tested in clinical trials as well as potential new avenues of investigation for the future.
Collapse
|
31
|
Orlandi A, Pagani F, Avitabile D, Bonanno G, Scambia G, Vigna E, Grassi F, Eusebi F, Fucile S, Pesce M, Capogrossi MC. Functional properties of cells obtained from human cord blood CD34+ stem cells and mouse cardiac myocytes in coculture. Am J Physiol Heart Circ Physiol 2008; 294:H1541-9. [PMID: 18223188 DOI: 10.1152/ajpheart.01285.2007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Prior in vitro studies suggested that different types of hematopoietic stem cells may differentiate into cardiomyocytes. The present work examined whether human CD34(+) cells from the human umbilical cord blood (hUCB), cocultured with neonatal mouse cardiomyocytes, acquire the functional properties of myocardial cells and express human cardiac genes. hUCB CD34(+) cells were cocultured onto cardiomyocytes following an infection with a lentivirus-encoding enhanced green fluorescent protein (EGFP). After 7 days, mononucleated EGFP(+) cells were tested for their electrophysiological features by patch clamp and for cytosolic [Ca(2+)] ([Ca(2+)](i)) homeostasis by [Ca(2+)](i) imaging of X-rhod1-loaded cells. Human Nkx2.5 and GATA-4 expression was examined in cocultured cell populations by real-time RT-PCR. EGFP(+) cells were connected to surrounding cells by gap junctions, acquired electrophysiological properties similar to those of cardiomyocytes, and showed action potential-associated [Ca(2+)](i) transients. These cells also exhibited spontaneous sarcoplasmic reticulum [Ca(2+)](i) oscillations and the associated membrane potential depolarization. However, RT-PCR of both cell populations showed no upregulation of human-specific cardiac genes. In conclusion, under our experimental conditions, hUCB CD34(+) cells cocultured with murine cardiomyocytes formed cells that exhibited excitation-contraction coupling features similar to those of cardiomyocytes. However, the expression of human-specific cardiac genes was undetectable by RT-PCR.
Collapse
Affiliation(s)
- Alessia Orlandi
- Laboratorio di Biologia Vascolare e Terapia Genica, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Arom KV, Ruengsakulrach P, Jotisakulratana V. Efficacy of Intramyocardial Injection of Angiogenic Cell Precursors for Ischemic Cardiomyopathy. INNOVATIONS-TECHNOLOGY AND TECHNIQUES IN CARDIOTHORACIC AND VASCULAR SURGERY 2008. [DOI: 10.1177/155698450800300109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Kitipan V. Arom
- Division of Cardiovascular Surgery, Bangkok Heart Hospital, Bangkok, Thailand
| | | | | |
Collapse
|
33
|
Eve DJ, Marty PJ, McDermott RJ, Klasko SK, Sanberg PR. Stem Cell Research and Health Education. AMERICAN JOURNAL OF HEALTH EDUCATION 2008; 39:167-179. [PMID: 19672471 PMCID: PMC2723780 DOI: 10.1901/jaba.2008.39-167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Stem cells are being touted as the greatest discovery for the potential treatment of a myriad of diseases in the new millennium, but there is still much research to be done before it will be known whether they can live up to this description. There is also an ethical debate over the production of one of the most valuable types of stem cell: the embryonic form. Consequently, there is public confusion over the benefits currently being derived from the use of stem cells and what can potentially be expected from their use in the future. The health educator's role is to give an unbiased account of the current state of stem cell research. This paper provides the groundwork by discussing the types of cells currently identified, their potential use, and some of the political and ethical pitfalls resulting from such use.
Collapse
Affiliation(s)
- David J. Eve
- David Eve is an instructor at the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd. (MDC 078), Tampa, FL 33612; E-mail: . Phillip J. Marty and Robert J. McDermott are professors in the Department of Community and Family Health, University of South Florida College of Public Health; E-mail: and . Stephen K. Klasko is vice president of USF Health and dean of the College of Medicine at the University of South Florida; E-mail: . Paul R. Sanberg is a distinguished university professor and director of the Center of Excellence for Aging and Brain Repair at the University of South Florida; E-mail:
| | - Phillip J. Marty
- David Eve is an instructor at the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd. (MDC 078), Tampa, FL 33612; E-mail: . Phillip J. Marty and Robert J. McDermott are professors in the Department of Community and Family Health, University of South Florida College of Public Health; E-mail: and . Stephen K. Klasko is vice president of USF Health and dean of the College of Medicine at the University of South Florida; E-mail: . Paul R. Sanberg is a distinguished university professor and director of the Center of Excellence for Aging and Brain Repair at the University of South Florida; E-mail:
| | - Robert J. McDermott
- David Eve is an instructor at the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd. (MDC 078), Tampa, FL 33612; E-mail: . Phillip J. Marty and Robert J. McDermott are professors in the Department of Community and Family Health, University of South Florida College of Public Health; E-mail: and . Stephen K. Klasko is vice president of USF Health and dean of the College of Medicine at the University of South Florida; E-mail: . Paul R. Sanberg is a distinguished university professor and director of the Center of Excellence for Aging and Brain Repair at the University of South Florida; E-mail:
| | - Stephen K. Klasko
- David Eve is an instructor at the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd. (MDC 078), Tampa, FL 33612; E-mail: . Phillip J. Marty and Robert J. McDermott are professors in the Department of Community and Family Health, University of South Florida College of Public Health; E-mail: and . Stephen K. Klasko is vice president of USF Health and dean of the College of Medicine at the University of South Florida; E-mail: . Paul R. Sanberg is a distinguished university professor and director of the Center of Excellence for Aging and Brain Repair at the University of South Florida; E-mail:
| | - Paul R. Sanberg
- David Eve is an instructor at the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd. (MDC 078), Tampa, FL 33612; E-mail: . Phillip J. Marty and Robert J. McDermott are professors in the Department of Community and Family Health, University of South Florida College of Public Health; E-mail: and . Stephen K. Klasko is vice president of USF Health and dean of the College of Medicine at the University of South Florida; E-mail: . Paul R. Sanberg is a distinguished university professor and director of the Center of Excellence for Aging and Brain Repair at the University of South Florida; E-mail:
| |
Collapse
|
34
|
Efficacy of intramyocardial injection of angiogenic cell precursors for ischemic cardiomyopathy: a case match study. INNOVATIONS-TECHNOLOGY AND TECHNIQUES IN CARDIOTHORACIC AND VASCULAR SURGERY 2008; 3:38-45. [PMID: 22436722 DOI: 10.1097/imi.0b013e31816755dd] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION : The objective of this study is to determine the efficacy of intramyocardial angiogenic cell precursors (ACPs) injection in ischemic cardiomyopathy (ICM). METHODS : Twenty-five ICM patients (cell group) underwent intramyocardial ACPs injection. Seventeen ICM patients (control group) treated by medical means were matched with cell group. There was no statistically significant difference between cell and control groups in relation to left ventricular ejection fraction (LVEF) and comorbidities. In the cell group, mean age was 58.4 ± 13.7 years. Mean LVEF was 26.1% ± 7.4%. New York Heart Association (NYHA) class was 2.9 ± 0.6. The ACPs were derived and expanded from autologous blood. The number of cells before injection was 27.4 ± 18.8 million cells. The cells were injected into the nonviable myocardium and hypokinetic segments in the cell group. RESULTS : There was no new ventricular arrhythmia. NYHA was improved by 0.9 ± 1.0 (P < 0.001) at 229.9 ± 98.8 days. Six-minute walk test and quality of life assessed by short form-36 improved in the cell group. LVEF was improved in 72% of patients (18 of 25). LVEF improved by 6.4 ± 9.9 points % (P = 0.003) at 290.4 ± 210.3 days. The percentage of infarction area decreased 21.9 ± 17.4 points % at 159 ± 54 days postoperatively. There was no significant improvement of NYHA and LVEF in the control group. CONCLUSIONS : For this efficacy study, the NYHA class, quality of life, and six-minute walk test were improved after cell transplantation. The LVEF was also significantly improved in the cell treated group.
Collapse
|
35
|
Affiliation(s)
- Luc Pardanaud
- Inserm U833, Chaire de Médecine Expérimentale, Collège de France, 11, place Marcelin Berthelot, 75005 Paris, France.
| |
Collapse
|
36
|
Fadini GP, Sartore S, Agostini C, Avogaro A. Significance of endothelial progenitor cells in subjects with diabetes. Diabetes Care 2007; 30:1305-13. [PMID: 17277037 DOI: 10.2337/dc06-2305] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Gian Paolo Fadini
- Department of Clinical and Experimental Medicine, University of Padova Medical School, Padova, Italy.
| | | | | | | |
Collapse
|
37
|
|
38
|
Zipori D. The stem state: the nature of normal stem cells and the relevance to cancer. Regen Med 2007; 2:29-30. [PMID: 24692888 DOI: 10.2217/17460751.2.1.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- D Zipori
- Department of Molecular Cell Biology, Weizmann Institute of Science , Rehovot, Israel
| |
Collapse
|