1
|
Taheri M, Tehrani HA, Dehghani S, Alibolandi M, Arefian E, Ramezani M. Nanotechnology and bioengineering approaches to improve the potency of mesenchymal stem cell as an off-the-shelf versatile tumor delivery vehicle. Med Res Rev 2024; 44:1596-1661. [PMID: 38299924 DOI: 10.1002/med.22023] [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: 12/08/2022] [Revised: 11/28/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
Targeting actionable mutations in oncogene-driven cancers and the evolution of immuno-oncology are the two prominent revolutions that have influenced cancer treatment paradigms and caused the emergence of precision oncology. However, intertumoral and intratumoral heterogeneity are the main challenges in both fields of precision cancer treatment. In other words, finding a universal marker or pathway in patients suffering from a particular type of cancer is challenging. Therefore, targeting a single hallmark or pathway with a single targeted therapeutic will not be efficient for fighting against tumor heterogeneity. Mesenchymal stem cells (MSCs) possess favorable characteristics for cellular therapy, including their hypoimmune nature, inherent tumor-tropism property, straightforward isolation, and multilineage differentiation potential. MSCs can be loaded with various chemotherapeutics and oncolytic viruses. The combination of these intrinsic features with the possibility of genetic manipulation makes them a versatile tumor delivery vehicle that can be used for in vivo selective tumor delivery of various chemotherapeutic and biological therapeutics. MSCs can be used as biofactory for the local production of chemical or biological anticancer agents at the tumor site. MSC-mediated immunotherapy could facilitate the sustained release of immunotherapeutic agents specifically at the tumor site, and allow for the achievement of therapeutic concentrations without the need for repetitive systemic administration of high therapeutic doses. Despite the enthusiasm evoked by preclinical studies that used MSC in various cancer therapy approaches, the translation of MSCs into clinical applications has faced serious challenges. This manuscript, with a critical viewpoint, reviewed the preclinical and clinical studies that have evaluated MSCs as a selective tumor delivery tool in various cancer therapy approaches, including gene therapy, immunotherapy, and chemotherapy. Then, the novel nanotechnology and bioengineering approaches that can improve the potency of MSC for tumor targeting and overcoming challenges related to their low localization at the tumor sites are discussed.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
2
|
Baig MS, Ahmad A, Pathan RR, Mishra RK. Precision Nanomedicine with Bio-Inspired Nanosystems: Recent Trends and Challenges in Mesenchymal Stem Cells Membrane-Coated Bioengineered Nanocarriers in Targeted Nanotherapeutics. J Xenobiot 2024; 14:827-872. [PMID: 39051343 PMCID: PMC11270309 DOI: 10.3390/jox14030047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/09/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
In the recent past, the formulation and development of nanocarriers has been elaborated into the broader fields and opened various avenues in their preclinical and clinical applications. In particular, the cellular membrane-based nanoformulations have been formulated to surpass and surmount the limitations and restrictions associated with naïve or free forms of therapeutic compounds and circumvent various physicochemical and immunological barriers including but not limited to systemic barriers, microenvironmental roadblocks, and other cellular or subcellular hinderances-which are quite heterogeneous throughout the diseases and patient cohorts. These limitations in drug delivery have been overcome through mesenchymal cells membrane-based precision therapeutics, where these interventions have led to the significant enhancements in therapeutic efficacies. However, the formulation and development of nanocarriers still focuses on optimization of drug delivery paradigms with a one-size-fits-all resolutions. As mesenchymal stem cell membrane-based nanocarriers have been engineered in highly diversified fashions, these are being optimized for delivering the drug payloads in more and better personalized modes, entering the arena of precision as well as personalized nanomedicine. In this Review, we have included some of the advanced nanocarriers which have been designed and been utilized in both the non-personalized as well as precision applicability which can be employed for the improvements in precision nanotherapeutics. In the present report, authors have focused on various other aspects of the advancements in stem cells membrane-based nanoparticle conceptions which can surmount several roadblocks and barriers in drug delivery and nanomedicine. It has been suggested that well-informed designing of these nanocarriers will lead to appreciable improvements in the therapeutic efficacy in therapeutic payload delivery applications. These approaches will also enable the tailored and customized designs of MSC-based nanocarriers for personalized therapeutic applications, and finally amending the patient outcomes.
Collapse
Affiliation(s)
- Mirza Salman Baig
- Anjuman-I-Islam Kalsekar Technical Campus School of Pharmacy, Sector-16, Near Thana Naka, Khandagao, New Panvel, Navi Mumbai 410206, Maharashtra, India;
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, Foothills Medical Centre, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - Rakesh Kumar Mishra
- School of Health Sciences and Technology, University of Petroleum and Energy Studies (UPES), Bidholi, Dehradun 248007, Uttarakhand, India;
| |
Collapse
|
3
|
Derakhshandeh K, Ghalaei PM, Aryaeinejad S, Hoseini SA. Wheat germ agglutinin conjugated chitosan nanoparticles for gemcitabine delivery in MCF-7 cells; synthesis, characterisation and in vitro cytotoxicity studies. J Cancer Res Ther 2024; 20:167-175. [PMID: 38554316 DOI: 10.4103/jcrt.jcrt_1583_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/11/2022] [Indexed: 04/01/2024]
Abstract
OBJECTIVE AND AIM Numerous clinical trials indicated combination regimens containing gemcitabine could extend progression-free survival of breast cancer patients without increasing the incidence of serious adverse effects. Orally administered gemcitabine is being metabolized by enzymes present in intestinal cells rapidly; thereupon, the current study was aimed to preparing, optimizing, and evaluating cytotoxicity of wheat germ agglutinin conjugated gemcitabine-chitosan nanoparticles (WGA-Gem-CNPs) in MCF-7 and HEK293 cells and to determining their cellular uptake by Caco-2 cells. METHODS Gem-CNPs were prepared by Ionic Gelation method and optimum formulation was implied for WGA conjugation optimisation. Nanoparticles formation was approved by FTIR and DSC analyses; then particles were characterized by DLS and release profile was prepared. MTT assay was performed in MCF-7 and HEK293. RESULTS Optimized Gem-CNPs and WGA-Gem-CNPs particle size were estimated as 126.6 ± 21.8 and 144.8 ± 36.1 nm, respectively. WGA conjugation efficacy was calculated as 50.98 ± 2.32 percent and encapsulation efficiency in WGA-Gem-CNPs was 69.44 ± 3.41 percent. Three-hour Caco-2 cellular uptake from Gem-CNPs and WGA-Gem-CNPs were estimated as averagely 3.5 and 4.5 folds higher than free drug, respectively. Gem-CNPs and WGA-Gem-CNPs reduced IC50 in MCF-7 cells by 2 and 2.5 folds, respectively; such decrease for HEK293 cells was as much as 2.4 and 6.3 folds, in same order. CONCLUSION Demonstrated significant in vitro uptake of WGA-Gem-CNPs and cytotoxicity might be considered for more studies as a potential carrier for oral delivery of gemcitabine.
Collapse
Affiliation(s)
- Katayoun Derakhshandeh
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | | | | |
Collapse
|
4
|
Astrelina TA, Brunchukov VA, Kodina GE, Bubenshchikov VB, Larenkov AA, Lunev AS, Petrosova KA, Rastorgueva AA, Kobzeva IV, Usupzhanova DY, Nikitina VA, Malsagova KA, Kulikova LI, Samoilov AS, Pustovoyt VI. Biodistribution of Mesenchymal Stromal Cells Labeled with [ 89Zr]Zr-Oxine in Local Radiation Injuries in Laboratory Animals. Molecules 2023; 28:7169. [PMID: 37894647 PMCID: PMC10609482 DOI: 10.3390/molecules28207169] [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: 08/14/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Tracking the migration pathways of living cells after their introduction into a patient's body is a topical issue in the field of cell therapy. Questions related to studying the possibility of long-term intravital biodistribution of mesenchymal stromal cells in the body currently remain open. METHODS Forty-nine laboratory animals were used in the study. Modeling of local radiation injuries was carried out, and the dynamics of the distribution of mesenchymal stromal cells labeled with [89Zr]Zr-oxine in the rat body were studied. RESULTS the obtained results of the labelled cell distribution allow us to assume that this procedure could be useful for visualization of local radiation injury using positron emission tomography. However, further research is needed to confirm this assumption. CONCLUSIONS intravenous injection leads to the initial accumulation of cells in the lungs and their subsequent redistribution to the liver, spleen, and kidneys. When locally injected into tissues, mesenchymal stromal cells are not distributed systemically in significant quantities.
Collapse
Affiliation(s)
- Tatiana A. Astrelina
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Vitaliy A. Brunchukov
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Galina E. Kodina
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Viktor B. Bubenshchikov
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Anton A. Larenkov
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Aleksandr S. Lunev
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Kristina A. Petrosova
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Anna A. Rastorgueva
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Irina V. Kobzeva
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Daria Y. Usupzhanova
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Victoria A. Nikitina
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | | | - Ludmila I. Kulikova
- Institute of Biomedical Chemistry, Biobanking Group, 119121 Moscow, Russia;
- Institute of Mathematical Problems of Biology RAS—The Branch of Keldysh Institute of Applied Mathematics of Russian Academy of Sciences, 142290 Pushchino, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 119991 Pushchino, Russia
| | - Alexander S. Samoilov
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Vasiliy I. Pustovoyt
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| |
Collapse
|
5
|
Yang Y, Yang Y, Liu D, Wang Y, Lu M, Zhang Q, Huang J, Li Y, Ma T, Yan F, Zheng H. In-vivo programmable acoustic manipulation of genetically engineered bacteria. Nat Commun 2023; 14:3297. [PMID: 37280199 DOI: 10.1038/s41467-023-38814-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 05/15/2023] [Indexed: 06/08/2023] Open
Abstract
Acoustic tweezers can control target movement through the momentum interaction between an acoustic wave and an object. This technology has advantages over optical tweezers for in-vivo cell manipulation due to its high tissue penetrability and strong acoustic radiation force. However, normal cells are difficult to acoustically manipulate because of their small size and the similarity between their acoustic impedance and that of the medium. In this study, we use the heterologous expression of gene clusters to generate genetically engineered bacteria that can produce numerous sub-micron gas vesicles in the bacterial cytoplasm. We show that the presence of the gas vesicles significantly enhances the acoustic sensitivity of the engineering bacteria, which can be manipulated by ultrasound. We find that by employing phased-array-based acoustic tweezers, the engineering bacteria can be trapped into clusters and manipulated in vitro and in vivo via electronically steered acoustic beams, enabling the counter flow or on-demand flow of these bacteria in the vasculature of live mice. Furthermore, we demonstrate that the aggregation efficiency of engineering bacteria in a tumour is improved by utilizing this technology. This study provides a platform for the in-vivo manipulation of live cells, which will promote the progress of cell-based biomedical applications.
Collapse
Affiliation(s)
- Ye Yang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- Shenzhen College of Advanced Technology, University of the Chinese Academy of Sciences, 100049, Beijing, China
| | - Yaozhang Yang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- Shenzhen Bay Laboratory, 518132, Shenzhen, China
| | - Dingyuan Liu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Yuanyuan Wang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Minqiao Lu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Qi Zhang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Jiqing Huang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Yongchuan Li
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Teng Ma
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
- Shenzhen College of Advanced Technology, University of the Chinese Academy of Sciences, 100049, Beijing, China.
| | - Fei Yan
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
- Shenzhen College of Advanced Technology, University of the Chinese Academy of Sciences, 100049, Beijing, China.
| | - Hairong Zheng
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
- Shenzhen College of Advanced Technology, University of the Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
6
|
Abstract
The homeostasis of cellular activities is essential for the normal functioning of living organisms. Hence, the ability to regulate the fates of cells is of great significance for both fundamental chemical biology studies and therapeutic development. Despite the notable success of small-molecule drugs that normally act on cellular protein functions, current clinical challenges have highlighted the use of macromolecules to tune cell function for improved therapeutic outcomes. As a class of hybrid biomacromolecules gaining rapidly increasing attention, protein conjugates have exhibited great potential as versatile tools to manipulate cell function for therapeutic applications, including cancer treatment, tissue engineering, and regenerative medicine. Therefore, recent progress in the design and assembly of protein conjugates used to regulate cell function is discussed in this review. The protein conjugates covered here are classified into three different categories based on their mechanisms of action and relevant applications: (1) regulation of intercellular interactions; (2) intervention in intracellular biological pathways; (3) termination of cell proliferation. Within each genre, a variety of protein conjugate scaffolds are discussed, which contain a diverse array of grafted molecules, such as lipids, oligonucleotides, synthetic polymers, and small molecules, with an emphasis on their conjugation methodologies and potential biomedical applications. While the current generation of protein conjugates is focused largely on delivery, the next generation is expected to address issues of site-specific conjugation, in vivo stability, controllability, target selectivity, and biocompatibility.
Collapse
Affiliation(s)
- Yiao Wang
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carston R Wagner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mark D Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
7
|
Cell-based drug delivery systems and their in vivo fate. Adv Drug Deliv Rev 2022; 187:114394. [PMID: 35718252 DOI: 10.1016/j.addr.2022.114394] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/17/2022] [Accepted: 06/07/2022] [Indexed: 11/22/2022]
Abstract
Cell-based drug delivery systems (DDSs) have received attention recently because of their unique biological properties and self-powered functions, such as excellent biocompatibility, low immunogenicity, long circulation time, tissue-homingcharacteristics, and ability to cross biological barriers. A variety of cells, including erythrocytes, stem cells, and lymphocytes, have been explored as functional vectors for the loading and delivery of various therapeutic payloads (e.g., small-molecule and nucleic acid drugs) for subsequent disease treatment. These cell-based DDSs have their own unique in vivo fates, which are attributed to various factors, including their biological properties and functions, the loaded drugs and loading process, physiological and pathological circumstances, and the body's response to these carrier cells, which result in differences in drug delivery efficiency and therapeutic effect. In this review, we summarize the main cell-based DDSs and their biological properties and functions, applications in drug delivery and disease treatment, and in vivo fate and influencing factors. We envision that the unique biological properties, combined with continuing research, will enable development of cell-based DDSs as friendly drug vectors for the safe, effective, and even personalized treatment of diseases.
Collapse
|
8
|
Sanchez-Diaz M, Quiñones-Vico MI, Sanabria de la Torre R, Montero-Vílchez T, Sierra-Sánchez A, Molina-Leyva A, Arias-Santiago S. Biodistribution of Mesenchymal Stromal Cells after Administration in Animal Models and Humans: A Systematic Review. J Clin Med 2021; 10:jcm10132925. [PMID: 34210026 PMCID: PMC8268414 DOI: 10.3390/jcm10132925] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal Stromal Cells (MSCs) are of great interest in cellular therapy. Different routes of administration of MSCs have been described both in pre-clinical and clinical reports. Knowledge about the fate of the administered cells is critical for developing MSC-based therapies. The aim of this review is to describe how MSCs are distributed after injection, using different administration routes in animal models and humans. A literature search was performed in order to consider how MSCs distribute after intravenous, intraarterial, intramuscular, intraarticular and intralesional injection into both animal models and humans. Studies addressing the biodistribution of MSCs in “in vivo” animal models and humans were included. After the search, 109 articles were included in the review. Intravenous administration of MSCs is widely used; it leads to an initial accumulation of cells in the lungs with later redistribution to the liver, spleen and kidneys. Intraarterial infusion bypasses the lungs, so MSCs distribute widely throughout the rest of the body. Intramuscular, intraarticular and intradermal administration lack systemic biodistribution. Injection into various specific organs is also described. Biodistribution of MSCs in animal models and humans appears to be similar and depends on the route of administration. More studies with standardized protocols of MSC administration could be useful in order to make results homogeneous and more comparable.
Collapse
Affiliation(s)
- Manuel Sanchez-Diaz
- Dermatology Department, Hospital Universitario Virgen de las Nieves, IBS Granada, 18014 Granada, Spain; (M.S.-D.); (T.M.-V.); (A.M.-L.); (S.A.-S.)
| | - Maria I. Quiñones-Vico
- Cellular Production Unit, Hospital Universitario Virgen de las Nieves, IBS Granada, 18014 Granada, Spain; (R.S.d.l.T.); (A.S.-S.)
- Correspondence:
| | - Raquel Sanabria de la Torre
- Cellular Production Unit, Hospital Universitario Virgen de las Nieves, IBS Granada, 18014 Granada, Spain; (R.S.d.l.T.); (A.S.-S.)
| | - Trinidad Montero-Vílchez
- Dermatology Department, Hospital Universitario Virgen de las Nieves, IBS Granada, 18014 Granada, Spain; (M.S.-D.); (T.M.-V.); (A.M.-L.); (S.A.-S.)
| | - Alvaro Sierra-Sánchez
- Cellular Production Unit, Hospital Universitario Virgen de las Nieves, IBS Granada, 18014 Granada, Spain; (R.S.d.l.T.); (A.S.-S.)
| | - Alejandro Molina-Leyva
- Dermatology Department, Hospital Universitario Virgen de las Nieves, IBS Granada, 18014 Granada, Spain; (M.S.-D.); (T.M.-V.); (A.M.-L.); (S.A.-S.)
| | - Salvador Arias-Santiago
- Dermatology Department, Hospital Universitario Virgen de las Nieves, IBS Granada, 18014 Granada, Spain; (M.S.-D.); (T.M.-V.); (A.M.-L.); (S.A.-S.)
- Cellular Production Unit, Hospital Universitario Virgen de las Nieves, IBS Granada, 18014 Granada, Spain; (R.S.d.l.T.); (A.S.-S.)
- School of Medicine, University of Granada, 18014 Granada, Spain
| |
Collapse
|
9
|
Mandel I, Farkasdi S, Varga G, Nagy ÁK. Comparative Evaluation of Two Hyaluronic Acid Gel Products for the Treatment of Interdental Papillary Defects. Acta Stomatol Croat 2020; 54:227-237. [PMID: 33132386 PMCID: PMC7586896 DOI: 10.15644/asc54/3/1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objectives The aim was to investigate the efficacy of single injections of two different hyaluronic acid products, Flex Barrier and Revident, in reducing the size of black triangles to treat Nordland-Tarnow Class I and II recessions. Materials and Methods Forty adult patients were recruited with at least two upper and two lower interdental papilla defects in the front region between canine teeth. According to the Nordland-Tarnow classification of papillary defects, both Class I and Class II recessions were included in the investigation. Patients were randomly assigned to experimental groups to receive single injections of two different hyaluronic acid products, either Flex Barrier or Revident. The untreated sites served as controls. Photographs were taken before and immediately after the treatment, and again after one week and one month. To determine the size of the black triangles, Image J software was used. For statistical analysis, a mixed-design ANOVA was applied. Results Both Flex Barrier and Revident significantly decreased the size of the treated defects immediately after the treatment and also one week later (p<0.001). The beneficial effect of Revident lasted longer than Flex Barrier as it remained significant even after one month in Revident-treated patients, however, not in the Flex Barrier-treated group. Furthermore, Nordland-Tarnow Class I lesions generally showed a greater improvement than Class II lesions. Conclusion In this proof-of-concept, randomized clinical trial we have demonstrated the clinical applicability of both Flex Barrier and Revident, although Revident gave longer-lasting improvements than Flex Barrier. Further trials are needed to optimize multiple-application protocols for treating gingival black triangles.
Collapse
Affiliation(s)
- Iván Mandel
- - University of Pécs, Medical School, Department of Dentistry, Oral and Maxillofacial Surgery, Pécs, Hungary
| | - Sándor Farkasdi
- - Semmelweis University, Faculty of Dentistry, Department of Oral Biology, Budapest, Hungary
| | - Gábor Varga
- - Semmelweis University, Faculty of Dentistry, Department of Oral Biology, Budapest, Hungary
| | - Ákos Károly Nagy
- - University of Pécs, Medical School, Department of Dentistry, Oral and Maxillofacial Surgery, Pécs, Hungary
| |
Collapse
|
10
|
Hayes AJ, Melrose J. Glycosaminoglycan and Proteoglycan Biotherapeutics in Articular Cartilage Protection and Repair Strategies: Novel Approaches to Visco‐supplementation in Orthobiologics. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Anthony J. Hayes
- Bioimaging Research HubCardiff School of BiosciencesCardiff University Cardiff CF10 3AX Wales UK
| | - James Melrose
- Graduate School of Biomedical EngineeringUNSW Sydney Sydney NSW 2052 Australia
- Raymond Purves Bone and Joint Research LaboratoriesKolling Institute of Medical ResearchRoyal North Shore Hospital and The Faculty of Medicine and HealthUniversity of Sydney St. Leonards NSW 2065 Australia
- Sydney Medical SchoolNorthernRoyal North Shore HospitalSydney University St. Leonards NSW 2065 Australia
| |
Collapse
|
11
|
Yun L, Wu T, Liu R, Li K, Zhang M. Structural Variation and Microrheological Properties of a Homogeneous Polysaccharide from Wheat Germ. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:2977-2987. [PMID: 29350530 DOI: 10.1021/acs.jafc.7b04730] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A novel polysaccharide (WGP) was purified from crude wheat germ polysaccharide by Sephacryl S-500HRgel filtration. The molecular weight of WGP was determined as 4.89 × 106 Da and consisted of arabinose, xylose, glucose, and galactose. Methylation analysis and 1D/2D nuclear magnetic resonance was used to analyze the structural characterization of WGP. WGP was mainly a backbone composed of (1 → 4)-linked-β-d-Xylp (19.01%) and (1 → 3, 4)-linked-β-d-Xylp (26.27%) residues, which was branched of (1 → 5)-linked α-l-Araf (28.09%) and (1 → 3,6)-linked β-d-Galp (12.11%) with β-d-Glcp (14.52%) as terminal unit. The calculated values of Turbiscan stability indexes suggested that WGP (0.1-0.5 mg/mL) is a stable system. Microrheology results showed that WGP can form gel behavior when the concentration of WGP ranges from 0.1 to 3 mg/mL. Results of in vitro assays showed that WGP could cause the proliferation of RAW264.7 macrophages, upregulating the release of TNF-α and IL-8 in the lymphocytes.
Collapse
Affiliation(s)
- Liyuan Yun
- State Key Laboratory of Food Nutrition and Safety (Tianjin University of Science and Technology) , Key Laboratory of Food Nutrition and Safety, Ministry of Education , Tianjin 300457 , China
| | - Tao Wu
- State Key Laboratory of Food Nutrition and Safety (Tianjin University of Science and Technology) , Key Laboratory of Food Nutrition and Safety, Ministry of Education , Tianjin 300457 , China
| | - Rui Liu
- State Key Laboratory of Food Nutrition and Safety (Tianjin University of Science and Technology) , Key Laboratory of Food Nutrition and Safety, Ministry of Education , Tianjin 300457 , China
| | - Kun Li
- State Key Laboratory of Food Nutrition and Safety (Tianjin University of Science and Technology) , Key Laboratory of Food Nutrition and Safety, Ministry of Education , Tianjin 300457 , China
| | - Min Zhang
- State Key Laboratory of Food Nutrition and Safety (Tianjin University of Science and Technology) , Key Laboratory of Food Nutrition and Safety, Ministry of Education , Tianjin 300457 , China
| |
Collapse
|
12
|
Tsai TL, Wang HC, Hung CH, Lin PC, Lee YS, Chen HHW, Su WC. Wheat germ agglutinin-induced paraptosis-like cell death and protective autophagy is mediated by autophagy-linked FYVE inhibition. Oncotarget 2017; 8:91209-91222. [PMID: 29207637 PMCID: PMC5710917 DOI: 10.18632/oncotarget.20436] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 07/11/2017] [Indexed: 01/07/2023] Open
Abstract
Wheat germ agglutinin (WGA) is a lectin that specifically binds cell surface glycoproteins and disrupts nuclear pore complex function through its interaction with POM121. Our data indicate WGA induces paraptosis-like cell death without caspase activation. We observed the main features of paraptosis, including cytoplasmic vacuolation, endoplasmic reticulum dilation and increased ER stress, and the unfolded protein response in WGA-treated cervical carcinoma cells. Conversion of microtubule-associated protein I light chain 3 (LC3-I) into LC3-II and punctuate formation suggestive of autophagy were observed in WGA-treated cells. WGA-induced autophagy antagonized paraptosis in HeLa and CaSKi cells, which expressed autophagy-linked FYVE (Alfy) protein, but not in SiHa cells that did not express Alfy. Alfy knockdown in HeLa cells induced paraptosis-like cell death. These data indicate that WGA-induced cell death occurs through paraptosis and that autophagy may exert a protective effect. WGA treatment and Alfy inhibition could be an effective therapeutic strategy for apoptosis-resistant cervical cancer cells.
Collapse
Affiliation(s)
- Tsung Lin Tsai
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hao Chen Wang
- Insititue of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun Hua Hung
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Peng Chan Lin
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi San Lee
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Helen H W Chen
- Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wu Chou Su
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Insititue of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
13
|
Kim H, Jeong H, Han S, Beack S, Hwang BW, Shin M, Oh SS, Hahn SK. Hyaluronate and its derivatives for customized biomedical applications. Biomaterials 2017; 123:155-171. [DOI: 10.1016/j.biomaterials.2017.01.029] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/23/2016] [Accepted: 01/27/2017] [Indexed: 01/02/2023]
|