1
|
Chin SP, Marzuki M, Tai L, Mohamed Shahrehan NA, Ricky C, Fanty A, Salleh A, Low CT, Then KY, Hoe SLL, Cheong SK. Dynamic tracking of human umbilical cord mesenchymal stem cells (hUC-MSCs) following intravenous administration in mice model. Regen Ther 2024; 25:273-283. [PMID: 38314402 PMCID: PMC10834363 DOI: 10.1016/j.reth.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/07/2024] [Accepted: 01/18/2024] [Indexed: 02/06/2024] Open
Abstract
Introduction In the past decades, human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) have sparked interest in cellular therapy due to their immunomodulatory properties. Nevertheless, the fate of hUC-MSCs in the body remains poorly understood. This study aimed to investigate the biodistribution, homing and clearance of systemically administered hUC-MSCs in healthy BALB/c mice model. Methods hUC-MSCs were labelled with GFP-Luc2 protein, followed by characterisation with flow cytometry. Upon intravenous infusion of transduced hUC-MSCs into the healthy BALB/c mice, the cells were dynamically monitored through the bioluminescent imaging (BLI) approach. Results Transduction of hUC-MSCs with GFP-Luc2 not only preserved the characteristics of MSCs, but also allowed live monitoring of transduced cells in the mice model. Upon systemic administration, BLI showed that transduced hUC-MSCs first localised predominantly in the lungs of healthy BALB/c mice and mainly remained in the lungs for up to 3 days before eventually cleared from the body. At terminal sacrifice, plasma chemistry biomarkers remained unchanged except for C-peptide levels, which were significantly reduced in the hUC-MSCs group. Histopathological findings further revealed that hUC-MSCs infusion did not cause any adverse effects and toxicity to lung, liver and heart tissues. Conclusions Collectively, systemically administrated hUC-MSCs was safe and demonstrated dynamic homing capacity before eventually disappearing from the body.
Collapse
Affiliation(s)
- Sze-Piaw Chin
- Cytopeutics Sdn Bhd, Cyberjaya, Selangor, Malaysia
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long, Selangor, Malaysia
| | - Marini Marzuki
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, NIH, Setia Alam, Selangor, Malaysia
| | - Lihui Tai
- Cytopeutics Sdn Bhd, Cyberjaya, Selangor, Malaysia
| | | | - Christine Ricky
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, NIH, Setia Alam, Selangor, Malaysia
| | - Audrey Fanty
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, NIH, Setia Alam, Selangor, Malaysia
| | - Annas Salleh
- Department of Veterinary Laboratory Diagnosis, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Chui Thean Low
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, NIH, Setia Alam, Selangor, Malaysia
| | | | - Susan Ling Ling Hoe
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, NIH, Setia Alam, Selangor, Malaysia
| | - Soon Keng Cheong
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long, Selangor, Malaysia
| |
Collapse
|
2
|
Liu H, Sun R, Wang L, Chen X, Li G, Cheng Y, Zhai G, Bay BH, Yang F, Gu N, Guo Y, Fan H. Biocompatible Iron Oxide Nanoring-Labeled Mesenchymal Stem Cells: An Innovative Magnetothermal Approach for Cell Tracking and Targeted Stroke Therapy. ACS NANO 2022; 16:18806-18821. [PMID: 36278899 DOI: 10.1021/acsnano.2c07581] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Labeling stem cells with magnetic nanoparticles is a promising technique for in vivo tracking and magnetic targeting of transplanted stem cells, which is critical for improving the therapeutic efficacy of cell therapy. However, conventional endocytic labeling with relatively poor labeling efficiency and a short labeling lifetime has hindered the implementation of these innovative enhancements in stem-cell-mediated regenerative medicine. Herein, we describe an advanced magnetothermal approach to label mesenchymal stem cells (MSCs) efficiently by local induction of heat-enhanced membrane permeability for magnetic resonance imaging (MRI) tracking and targeted therapy of stroke, where biocompatible γ-phase, ferrimagnetic vortex-domain iron oxide nanorings (γ-FVIOs) with superior magnetoresponsive properties were used as a tracer. This approach facilitates a safe and efficient labeling of γ-FVIOs as high as 150 pg of Fe per cell without affecting the MSCs proliferation and differentiation, which is 3.44-fold higher than that by endocytosis labeling. Such a high labeling efficiency not only enables the ultrasensitive magnetic resonance imaging (MRI) detection of sub-10 cells and long-term tracking of transplanted MSCs over 10 weeks but also endows transplanted MSCs with a magnetic manipulation ability in vivo. A proof-of-concept study using a rat stroke model showed that the labeled MSCs facilitated MRI tracking and magnetic targeting for efficient replacement therapy with a significantly reduced dosage of 5 × 104 transplanted cells. The findings in this study have demonstrated the great potential of the magnetothermal approach as an efficient labeling technique for future clinical usage.
Collapse
Affiliation(s)
- Hanrui Liu
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu610041, China
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an710127, China
| | - Ran Sun
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Lei Wang
- Molecular Imaging Center, West China Hospital, Sichuan University, Chengdu610041, China
| | - Xiaoyong Chen
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an710127, China
| | - Galong Li
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an710127, China
- School of Medicine, Northwest University, Xi'an710069, China
| | - Yu Cheng
- Institute for Regenerative Medicine, The Institute for Biomedical Engineering & Nano Science, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai200092, China
| | - Gaohong Zhai
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an710127, China
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, 4 Medical Drive, MD10, 117594, Singapore
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing210009, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing210009, China
| | - Yingkun Guo
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Haiming Fan
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an710127, China
- School of Medicine, Northwest University, Xi'an710069, China
| |
Collapse
|
3
|
Garrigós MM, de Oliveira FA, Nucci MP, Nucci LP, Alves ADH, Dias OFM, Gamarra LF. How mesenchymal stem cell cotransplantation with hematopoietic stem cells can improve engraftment in animal models. World J Stem Cells 2022; 14:658-679. [PMID: 36157912 PMCID: PMC9453272 DOI: 10.4252/wjsc.v14.i8.658] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/27/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Bone marrow transplantation (BMT) can be applied to both hematopoietic and nonhematopoietic diseases; nonetheless, it still comes with a number of challenges and limitations that contribute to treatment failure. Bearing this in mind, a possible way to increase the success rate of BMT would be cotransplantation of mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) to improve the bone marrow niche and secrete molecules that enhance the hematopoietic engraftment.
AIM To analyze HSC and MSC characteristics and their interactions through cotransplantation in murine models.
METHODS We searched for original articles indexed in PubMed and Scopus during the last decade that used HSC and MSC cotransplantation and in vivo BMT in animal models while evaluating cell engraftment. We excluded in vitro studies or studies that involved graft versus host disease or other hematological diseases and publications in languages other than English. In PubMed, we initially identified 555 articles and after selection, only 12 were chosen. In Scopus, 2010 were identified, and six were left after the screening and eligibility process.
RESULTS Of the 2565 articles found in the databases, only 18 original studies met the eligibility criteria. HSC distribution by source showed similar ratios, with human umbilical cord blood or animal bone marrow being administered mainly with a dose of 1 × 107 cells by intravenous or intrabone routes. However, MSCs had a high prevalence of human donors with a variety of sources (umbilical cord blood, bone marrow, tonsil, adipose tissue or fetal lung), using a lower dose, mainly 106 cells and ranging 104 to 1.5 × 107 cells, utilizing the same routes. MSCs were characterized prior to administration in almost every experiment. The recipient used was mostly immunodeficient mice submitted to low-dose irradiation or chemotherapy. The main technique of engraftment for HSC and MSC cotransplantation evaluation was chimerism, followed by hematopoietic reconstitution and survival analysis. Besides the engraftment, homing and cellularity were also evaluated in some studies.
CONCLUSION The preclinical findings validate the potential of MSCs to enable HSC engraftment in vivo in both xenogeneic and allogeneic hematopoietic cell transplantation animal models, in the absence of toxicity.
Collapse
Affiliation(s)
- Murilo Montenegro Garrigós
- Hospital Israelita Albert Einstein, São Paulo 05652-900, São Paulo, Brazil
- Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, São Paulo, Brazil
| | | | - Mariana Penteado Nucci
- Hospital Israelita Albert Einstein, São Paulo 05652-900, São Paulo, Brazil
- LIM44-Hospital das Clínicas, Faculdade Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Leopoldo Penteado Nucci
- Centro Universitário do Planalto Central, Área Especial para Industria nº 02 Setor Leste - Gama-DF, Brasília 72445-020, Distrito Federal, Brazil
| | | | | | | |
Collapse
|
4
|
Optimization of Multimodal Nanoparticles Internalization Process in Mesenchymal Stem Cells for Cell Therapy Studies. Pharmaceutics 2022; 14:pharmaceutics14061249. [PMID: 35745821 PMCID: PMC9227698 DOI: 10.3390/pharmaceutics14061249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022] Open
Abstract
Considering there are several difficulties and limitations in labeling stem cells using multifunctional nanoparticles (MFNP), the purpose of this study was to determine the optimal conditions for labeling human bone marrow mesenchymal stem cells (hBM-MSC), aiming to monitor these cells in vivo. Thus, this study provides information on hBM-MSC direct labeling using multimodal nanoparticles in terms of concentration, magnetic field, and period of incubation while maintaining these cells’ viability and the homing ability for in vivo experiments. The cell labeling process was assessed using 10, 30, and 50 µg Fe/mL of MFNP, with periods of incubation ranging from 4 to 24 h, with or without a magnetic field, using optical microscopy, near-infrared fluorescence (NIRF), and inductively coupled plasma mass spectrometry (ICP-MS). After the determination of optimal labeling conditions, these cells were applied in vivo 24 h after stroke induction, intending to evaluate cell homing and improve NIRF signal detection. In the presence of a magnetic field and utilizing the maximal concentration of MFNP during cell labeling, the iron load assessed by NIRF and ICP-MS was four times higher than what was achieved before. In addition, considering cell viability higher than 98%, the recommended incubation time was 9 h, which corresponded to a 25.4 pg Fe/cell iron load (86% of the iron load internalized in 24 h). The optimization of cellular labeling for application in the in vivo study promoted an increase in the NIRF signal by 215% at 1 h and 201% at 7 h due to the use of a magnetized field during the cellular labeling process. In the case of BLI, the signal does not depend on cell labeling showing no significant differences between unlabeled or labeled cells (with or without a magnetic field). Therefore, the in vitro cellular optimized labeling process using magnetic fields resulted in a shorter period of incubation with efficient iron load internalization using higher MFNP concentration (50 μgFe/mL), leading to significant improvement in cell detection by NIRF technique without compromising cellular viability in the stroke model.
Collapse
|
5
|
Maric DM, Velikic G, Maric DL, Supic G, Vojvodic D, Petric V, Abazovic D. Stem Cell Homing in Intrathecal Applications and Inspirations for Improvement Paths. Int J Mol Sci 2022; 23:ijms23084290. [PMID: 35457107 PMCID: PMC9027729 DOI: 10.3390/ijms23084290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/26/2022] [Accepted: 04/01/2022] [Indexed: 02/04/2023] Open
Abstract
A transplanted stem cell homing is a directed migration from the application site to the targeted tissue. Intrathecal application of stem cells is their direct delivery to cerebrospinal fluid, which defines the homing path from the point of injection to the brain. In the case of neurodegenerative diseases, this application method has the advantage of no blood–brain barrier restriction. However, the homing efficiency still needs improvement and homing mechanisms elucidation. Analysis of current research results on homing mechanisms in the light of intrathecal administration revealed a discrepancy between in vivo and in vitro results and a gap between preclinical and clinical research. Combining the existing research with novel insights from cutting-edge biochips, nano, and other technologies and computational models may bridge this gap faster.
Collapse
Affiliation(s)
- Dusan M. Maric
- Department for Research and Development, Clinic Orto MD-Parks Dr Dragi Hospital, 21000 Novi Sad, Serbia;
- Faculty of Dentistry Pancevo, University Business Academy, 26000 Pancevo, Serbia
- Vincula Biotech Group, 11000 Belgrade, Serbia;
| | - Gordana Velikic
- Department for Research and Development, Clinic Orto MD-Parks Dr Dragi Hospital, 21000 Novi Sad, Serbia;
- Vincula Biotech Group, 11000 Belgrade, Serbia;
- Correspondence: (G.V.); (D.L.M.)
| | - Dusica L. Maric
- Department of Anatomy, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Correspondence: (G.V.); (D.L.M.)
| | - Gordana Supic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia; (G.S.); (D.V.)
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Danilo Vojvodic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia; (G.S.); (D.V.)
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Vedrana Petric
- Infectious Diseases Clinic, Clinical Center of Vojvodina, 21000 Novi Sad, Serbia;
- Department of Infectious Diseases, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Dzihan Abazovic
- Vincula Biotech Group, 11000 Belgrade, Serbia;
- Department for Regenerative Medicine, Biocell Hospital, 11000 Belgrade, Serbia
| |
Collapse
|
6
|
Mousaei Ghasroldasht M, Seok J, Park HS, Liakath Ali FB, Al-Hendy A. Stem Cell Therapy: From Idea to Clinical Practice. Int J Mol Sci 2022; 23:ijms23052850. [PMID: 35269990 PMCID: PMC8911494 DOI: 10.3390/ijms23052850] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/20/2022] [Accepted: 03/03/2022] [Indexed: 11/27/2022] Open
Abstract
Regenerative medicine is a new and promising mode of therapy for patients who have limited or no other options for the treatment of their illness. Due to their pleotropic therapeutic potential through the inhibition of inflammation or apoptosis, cell recruitment, stimulation of angiogenesis, and differentiation, stem cells present a novel and effective approach to several challenging human diseases. In recent years, encouraging findings in preclinical studies have paved the way for many clinical trials using stem cells for the treatment of various diseases. The translation of these new therapeutic products from the laboratory to the market is conducted under highly defined regulations and directives provided by competent regulatory authorities. This review seeks to familiarize the reader with the process of translation from an idea to clinical practice, in the context of stem cell products. We address some required guidelines for clinical trial approval, including regulations and directives presented by the Food and Drug Administration (FDA) of the United States, as well as those of the European Medicine Agency (EMA). Moreover, we review, summarize, and discuss regenerative medicine clinical trial studies registered on the Clinicaltrials.gov website.
Collapse
|
7
|
Nucci MP, Oliveira FA, Ferreira JM, Pinto YO, Alves AH, Mamani JB, Nucci LP, Valle NME, Gamarra LF. Effect of Cell Therapy and Exercise Training in a Stroke Model, Considering the Cell Track by Molecular Image and Behavioral Analysis. Cells 2022; 11:cells11030485. [PMID: 35159294 PMCID: PMC8834410 DOI: 10.3390/cells11030485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/12/2022] Open
Abstract
The goal of this study is to see how combining physical activity with cell treatment impacts functional recovery in a stroke model. Molecular imaging and multimodal nanoparticles assisted in cell tracking and longitudinal monitoring (MNP). The viability of mesenchymal stem cell (MSC) was determined using a 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay and bioluminescent image (BLI) after lentiviral transduction and MNP labeling. At random, the animals were divided into 5 groups (control-G1, and experimental G2-G5). The photothrombotic stroke induction was confirmed by local blood perfusion reduction and Triphenyltetrazolium chloride (TTC), and MSC in the G3 and G5 groups were implanted after 24 h, with BLI and near-infrared fluorescence image (NIRF) tracking these cells at 28 h, 2, 7, 14, and 28 days. During a 28-day period, the G5 also conducted physical training, whereas the G4 simply did the training. At 0, 7, 14, and 28 days, the animals were functionally tested using a cylinder test and a spontaneous motor activity test. MNP internalization in MSC was confirmed using brightfield and fluorescence microscopy. In relation to G1 group, only 3% of cell viability reduced. The G2–G5 groups showed more than 69% of blood perfusion reduction. The G5 group performed better over time, with a progressive recovery of symmetry and an increase of fast vertical movements. Up to 7 days, BLI and NIRF followed MSC at the damaged site, demonstrating a signal rise that could be connected to cell proliferation at the injury site during the acute phase of stroke. Local MSC therapy mixed with physical activity resulted in better results in alleviating motor dysfunction, particularly during the acute period. When it comes to neurorehabilitation, this alternative therapy could be a suitable fit.
Collapse
Affiliation(s)
- Mariana P. Nucci
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
- LIM44, Hospital das Clínicas da Faculdade Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Fernando A. Oliveira
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - João M. Ferreira
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - Yolanda O. Pinto
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - Arielly H. Alves
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - Javier B. Mamani
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - Leopoldo P. Nucci
- Centro Universitário do Planalto Central, Brasília 72445-020, Brazil;
| | - Nicole M. E. Valle
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - Lionel F. Gamarra
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
- Correspondence: ; Tel.: +55-11-2151-0243
| |
Collapse
|
8
|
Fan X, Wei H, Du J, Lu X, Wang L. Hypoxic preconditioning neural stem cell transplantation promotes spinal cord injury in rats by affecting transmembrane immunoglobulin domain-containing. Hum Exp Toxicol 2022; 41:9603271211066587. [PMID: 35243930 DOI: 10.1177/09603271211066587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To explore the effects of hypoxic preconditioning neural stem cell (P-NSC) transplantation on rats with spinal cord injury (SCI). METHODS After identification, the NSCs were treated with hypoxic preconditioning. The NSCs migration was detected by Transwell method. RT-qPCR was used to detect the mRNA levels of HIF-1α, CXCR4 in NSC. The secretion of representative neurotrophic factors (VEGF, HGF, and BDNF) was checked by Western blot. Forty-six SCI rats were randomly divided into three experimental groups: SCI group (PBS injection, n = 10); N-NSC group (NSC atmospheric normoxic pretreatment injection, n = 18); and P-NSC group (NSC 's hypoxic preconditioning injection, n = 18). The sham operation group was also included (rats underwent laminectomy but not SCI, n = 10). The recovery of hindlimb motor function was evaluated by BBB score. The level of spinal cord inflammation (IL-1β, TNF-α, and IL-6) was determined by ELISA. Western blot was used to detect the content of TMIGD1 and TMIGD3 in spinal cord. RESULTS Compared with the N-NSC group, the number of NSC-passing membranes in the P-NSC group increased with the increase of the culture time (p < 0.05). Compared with N-NSC, P-NSC had higher levels of VEGF, HGF, and BDNF after 1 week of culture (p < 0.05). The BBB score of the P-NSC group was significantly higher than that of the N-NSC group at 7 and 28 days (p < 0.05). Compared with the SCI group, the levels of TNF-α, IL-1β, and IL-6 were significantly reduced after NSC treatment, and the P-NSC group was lower than the N-NSC group (p < 0.05). Compared with the SCI group, the levels of TMIGD1 and TMIGD3 increased. Compared with the N-NSC group, and the levels of TMIGD1 and TMIGD3 increased in the P-NSC group (p < 0.05). CONCLUSION P-NSC administration could improve SCI injury, and the levels of TMIGD1 and TMIGD3.
Collapse
Affiliation(s)
- Xiaoguang Fan
- The Second Department of Spine Surgery, 519688Yantaishan Hospital, Yantai, China
| | - Hongchun Wei
- Department of Neurology, 117747the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Juan Du
- Department of Neurology, 519688Yantaishan Hospital, Yantai, China
| | - Xiuguo Lu
- Department of spine surgery, Yantai Yeda Hospital, Yantai, China
| | - Leisheng Wang
- The Second Department of Spine Surgery, 519688Yantaishan Hospital, Yantai, China
| |
Collapse
|
9
|
Oliveira FA, Nucci MP, Mamani JB, Alves AH, Rego GNA, Kondo AT, Hamerschlak N, Junqueira MS, de Souza LEB, Gamarra LF. Multimodal Tracking of Hematopoietic Stem Cells from Young and Old Mice Labeled with Magnetic-Fluorescent Nanoparticles and Their Grafting by Bioluminescence in a Bone Marrow Transplant Model. Biomedicines 2021; 9:biomedicines9070752. [PMID: 34209598 PMCID: PMC8301491 DOI: 10.3390/biomedicines9070752] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
This study proposes an innovative way to evaluate the homing and tracking of hematopoietic stem cells from young and old mice labeled with SPIONNIRF-Rh conjugated with two types of fluorophores (NIRF and Rhodamine), and their grafting by bioluminescence (BLI) in a bone marrow transplant (BMT) model. In an in vitro study, we isolated bone marrow mononuclear cells (BM-MNC) from young and old mice, and analyzed the physical-chemical characteristics of SPIONNIRF-Rh, their internalization, cell viability, and the iron quantification by NIRF, ICP-MS, and MRI. The in vivo study was performed in a BMT model to evaluate the homing, tracking, and grafting of young and old BM-MNC labeled with SPIONNIRF-Rh by NIRF and BLI, as well as the hematological reconstitution for 120 days. 5FU influenced the number of cells isolated mainly in young cells. SPIONNIRF-Rh had adequate characteristics for efficient internalization into BM-MNC. The iron load quantification by NIRF, ICP-MS, and MRI was in the order of 104 SPIONNIRF-Rh/BM-MNC. In the in vivo study, the acute NIRF evaluation showed higher signal intensity in the spinal cord and abdominal region, and the BLI evaluation allowed follow-up (11-120 days), achieving a peak of intensity at 30 days, which remained stable around 108 photons/s until the end. The hematologic evaluation showed similar behavior until 30 days and the histological results confirm that iron is present in almost all tissue evaluated. Our results on BM-MNC homing and tracking in the BMT model did not show a difference in migration or grafting of cells from young or old mice, with the hemogram analysis trending to differentiation towards the myeloid lineage in mice that received cells from old animals. The cell homing by NIRF and long term cell follow-up by BLI highlighted the relevance of the multimodal nanoparticles and combined techniques for evaluation.
Collapse
Affiliation(s)
- Fernando A. Oliveira
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Mariana P. Nucci
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
- LIM44—Hospital das Clínicas da Faculdade Medicina da Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | - Javier B. Mamani
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Arielly H. Alves
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Gabriel N. A. Rego
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Andrea T. Kondo
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Nelson Hamerschlak
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Mara S. Junqueira
- Center for Translational Research in Oncology, Cancer Institute of the State of Sao Paulo—ICESP, São Paulo 01246-000, SP, Brazil;
| | - Lucas E. B. de Souza
- Hemocentro de Ribeirão Preto, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14051-060, SP, Brazil;
| | - Lionel F. Gamarra
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (F.A.O.); (M.P.N.); (J.B.M.); (A.H.A.); (G.N.A.R.); (A.T.K.); (N.H.)
- Correspondence: ; Tel.: +55-11-2151-0243
| |
Collapse
|
10
|
Prospects of Therapeutic Target and Directions for Ischemic Stroke. Pharmaceuticals (Basel) 2021; 14:ph14040321. [PMID: 33916253 PMCID: PMC8065883 DOI: 10.3390/ph14040321] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 12/12/2022] Open
Abstract
Stroke is a serious, adverse neurological event and the third leading cause of death and disability worldwide. Most strokes are caused by a block in cerebral blood flow, resulting in neurological deficits through the death of brain tissue. Recombinant tissue plasminogen activator (rt-PA) is currently the only immediate treatment medication for stroke. The goal of rt-PA administration is to reduce the thrombus and/or embolism via thrombolysis; however, the administration of rt-PA must occur within a very short therapeutic timeframe (3 h to 6 h) after symptom onset. Components of the pathological mechanisms involved in ischemic stroke can be used as potential biomarkers in current treatment. However, none are currently under investigation in clinical trials; thus, further studies investigating biomarkers are needed. After ischemic stroke, microglial cells can be activated and release inflammatory cytokines. These cytokines lead to severe neurotoxicity via the overactivation of microglia in prolonged and lasting insults such as stroke. Thus, the balanced regulation of microglial activation may be necessary for therapy. Stem cell therapy is a promising clinical treatment strategy for ischemic stroke. Stem cells can increase the functional recovery of damaged tissue after post-ischemic stroke through various mechanisms including the secretion of neurotrophic factors, immunomodulation, the stimulation of endogenous neurogenesis, and neovascularization. To investigate the use of stem cell therapy for neurological diseases in preclinical studies, however, it is important to develop imaging technologies that are able to evaluate disease progression and to “chase” (i.e., track or monitor) transplanted stem cells in recipients. Imaging technology development is rapidly advancing, and more sensitive techniques, such as the invasive and non-invasive multimodal techniques, are under development. Here, we summarize the potential risk factors and biomarker treatment strategies, stem cell-based therapy and emerging multimodal imaging techniques in the context of stroke. This current review provides a conceptual framework for considering the therapeutic targets and directions for the treatment of brain dysfunctions, with a particular focus on ischemic stroke.
Collapse
|
11
|
Kumar R, Gulia K. The convergence of nanotechnology‐stem cell, nanotopography‐mechanobiology, and biotic‐abiotic interfaces: Nanoscale tools for tackling the top killer, arteriosclerosis, strokes, and heart attacks. NANO SELECT 2021. [DOI: 10.1002/nano.202000192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Rajiv Kumar
- NIET National Institute of Medical Science Rajasthan India
| | - Kiran Gulia
- Materials and Manufacturing School of Engineering University of Wolverhampton Wolverhampton England, UK
| |
Collapse
|
12
|
Deng P, Halmai J, Waldo JJ, Fink KD. Cell-Based Delivery Approaches for DNA-Binding Domains to the Central Nervous System. Curr Neuropharmacol 2021; 19:2125-2140. [PMID: 33998992 PMCID: PMC9185769 DOI: 10.2174/1570159x19666210517144044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/16/2021] [Accepted: 05/05/2021] [Indexed: 11/22/2022] Open
Abstract
Advancements in programmable DNA-Binding Proteins (DBDs) that target the genome, such as zinc fingers, transcription activator-like effectors, and Cas9, have broadened drug target design beyond traditional protein substrates. Effective delivery methodologies remain a major barrier in targeting the central nervous system. Currently, adeno-associated virus is the most wellvalidated delivery system for the delivery of DBDs towards the central nervous with multiple, ongoing clinical trials. While effective in transducing neuronal cells, viral delivery systems for DBDs remain problematic due to inherent viral packaging limits or immune responses that hinder translational potential. Direct administration of DBDs or encapsulation in lipid nanoparticles may provide alternative means towards delivering gene therapies into the central nervous system. This review will evaluate the strengths and limitations of current DBD delivery strategies in vivo. Furthermore, this review will discuss the use of adult stem cells as a putative delivery vehicle for DBDs and the potential advantages that these systems have over previous methodologies.
Collapse
Affiliation(s)
- Peter Deng
- Department of Neurology, Stem Cell Program and Gene Therapy Center, UC Davis Medical Center, Sacramento, CA, USA
| | - Julian Halmai
- Department of Neurology, Stem Cell Program and Gene Therapy Center, UC Davis Medical Center, Sacramento, CA, USA
| | - Jennifer J. Waldo
- Department of Neurology, Stem Cell Program and Gene Therapy Center, UC Davis Medical Center, Sacramento, CA, USA
| | - Kyle D. Fink
- Department of Neurology, Stem Cell Program and Gene Therapy Center, UC Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|