1
|
Dominguez-Pinilla N, González-Granado LI, Gonzaga A, López Diaz M, Castellano Yáñez C, Aymerich C, Freire X, Ordoñez O, Diaz de Atauri ÁG, Albi Rodríguez MS, Martínez López E, Iñiguez R, Serrano Garrote O, Frontiñán AC, Andreu E, Gutierrez-Vilchez AM, Anton-Bonete M, Martinez-Navarrete G, Castillo-Flores N, Prat-Vidal C, Blanco M, Morante Valverde R, Fernandez E, Querol S, Hernández-Blasco LM, Belda-Hofheinz S, Soria B. Consecutive intrabronchial administration of Wharton's jelly-derived mesenchymal stromal cells in ECMO-supported pediatric patients with end-stage interstitial lung disease: a safety and feasibility study (CIBA method). Stem Cell Res Ther 2025; 16:164. [PMID: 40188166 PMCID: PMC11972491 DOI: 10.1186/s13287-025-04289-3] [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: 01/29/2025] [Accepted: 03/19/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Patients ineligible for lung transplant with end-stage Interstitial Lung Disease (ILD) on Extra-Corporeal Membrane Oxygenation (ECMO) face an appalling prognosis with limited therapeutic options. Due to the beneficial effect of Mesenchymal Stromal Cells (MSC) on inflammatory, immunological and infectious diseases, cell therapy has been proposed as an option, but administration is hampered by the ECMO. METHODS Cryopreserved Wharton-jelly derived MSC (WJ-MSC) were conveniently diluted and directly applied consecutively on each lobule (5,1 ml = 107 cells) at a continuous slow rate infused over one hour via flexible bronchoscopy (Consecutive IntraBronchial Administration method, CIBA method). RESULTS Intrabronchial administration of MSC to a patient on ECMO was well tolerated by the patient even though it did not reverse the patient's ILD. This manuscript presents preliminary evidence from ongoing clinical trials program on Cell Therapy of Inflammatory, Immune and Infectious Diseases and, to our knowledge, is the first report of intrabronchial administration of MSC in a paediatric ECMO patient with ILD. Even more, MSC administered by this method do not reach the systemic circulation and do get blocked on ECMO membrane. CONCLUSIONS Direct intrabronchial administration of MSC in a patient on ECMO is feasible and safe, and may be a new avenue to be assayed in ECMO patients with inflammatory, immunological and infectious diseases of the lung.
Collapse
Affiliation(s)
| | | | - Aitor Gonzaga
- Institute for Health and Biomedical Research (ISABIAL), Dr. Balmis General and University Hospital, Alicante, Spain
- Institute of Bioengineering-University Miguel Hernández, Elche, Spain
| | | | | | - Clara Aymerich
- Paediatric Intensive Care Unit, Hospital 12 de Octubre, Madrid, Spain
| | - Xabier Freire
- Paediatric Intensive Care Unit, Hospital 12 de Octubre, Madrid, Spain
| | - Olga Ordoñez
- Paediatric Intensive Care Unit, Hospital 12 de Octubre, Madrid, Spain
| | | | | | | | | | | | | | - Etelvina Andreu
- Institute for Health and Biomedical Research (ISABIAL), Dr. Balmis General and University Hospital, Alicante, Spain
- Dept. Applied Physics, University Miguel Hernández Elche, Elche, Spain
| | - Ana María Gutierrez-Vilchez
- Institute of Bioengineering-University Miguel Hernández, Elche, Spain
- Dept. of Pharmacology, Pediatrics and Organic Chemistry, University Miguel Hernández, Elche, Spain
| | | | - Gema Martinez-Navarrete
- Institute of Bioengineering-University Miguel Hernández, Elche, Spain
- Dept. Histology and Anatomy, Faculty of Medicine, University Miguel Hernandez, Elche, Spain
| | | | | | | | | | - Eduardo Fernandez
- Institute of Bioengineering-University Miguel Hernández, Elche, Spain
- Dept. Histology and Anatomy, Faculty of Medicine, University Miguel Hernandez, Elche, Spain
- CIBER of Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain
| | | | - Luis Manuel Hernández-Blasco
- Institute for Health and Biomedical Research (ISABIAL), Dr. Balmis General and University Hospital, Alicante, Spain
- Pneumology Service, Dr Balmis General and University Hospital, Alicante, Spain
| | | | - Bernat Soria
- Institute for Health and Biomedical Research (ISABIAL), Dr. Balmis General and University Hospital, Alicante, Spain.
- Institute of Bioengineering-University Miguel Hernández, Elche, Spain.
- CIBER of Diabetes and Metabolic Diseases, CIBERDEM, Madrid, Spain.
| |
Collapse
|
2
|
Giesen M, Fleck E, Scheele J, Hartmann TN, Henschler R. Rap1 Guanosine Triphosphate Hydrolase (GTPase) Regulates Shear Stress-Mediated Adhesion of Mesenchymal Stromal Cells. BIOLOGY 2025; 14:96. [PMID: 39857326 PMCID: PMC11762871 DOI: 10.3390/biology14010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Intravenously transplanted mesenchymal stromal cells (MSCs) have been shown to interact with endothelial cells and to migrate to tissues. However, intracellular signals regulating MSC migration are still incompletely understood. Here, we analyzed the role of Rap1 GTPase in the migration of human bone marrow-derived MSCs in vitro and in short-term homing in mice in vivo. MSCs expressed both Rap1A and Rap1B mRNAs, which were downregulated after treatment with siRNA against Rap1A and/or B. In a flow chamber model with pre-established human umbilical vein endothelial cells (HUVECs), Rap1A/B downregulated MSCs interacted for longer distances before arrest, indicating adhesion defects. CXCL12-induced adhesion of MSCs on immobilized Vascular Cell Adhesion Molecule (VCAM)-1 was also decreased after the downregulation of Rap1A, Rap1B, or both, as was CXCL12-induced transwell migration. In a competitive murine short-term homing model with i.v. co-injection of Rap1A+B siRNA-treated and control MSCs that were labeled with PKH 26 and PKH 67 fluorescent dyes, the Rap1A+B siRNA-treated MSCs were detected at increased frequencies in blood, liver, and spleen compared to control MSCs. Thus, Rap1 GTPase modulates the adhesion and migration of MSCs in vitro and may increase the bio-availability of i.v.-transplanted MSCs in tissues in a murine model.
Collapse
Affiliation(s)
- Melanie Giesen
- Institute for Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Service, Clinics of the Goethe University Frankfurt (Main), 60528 Frankfurt am Main, Germany; (M.G.); (E.F.)
| | - Erika Fleck
- Institute for Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Service, Clinics of the Goethe University Frankfurt (Main), 60528 Frankfurt am Main, Germany; (M.G.); (E.F.)
| | - Jürgen Scheele
- Department of Medicine I, Medical Center University of Freiburg and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (J.S.); (T.N.H.)
| | - Tanja Nicole Hartmann
- Department of Medicine I, Medical Center University of Freiburg and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (J.S.); (T.N.H.)
| | - Reinhard Henschler
- Institute for Transfusion Medicine, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
3
|
Chiang BJ, Mao SH, Chen TS, Chung SD, Chien CT. Adipose stem-cell-derived microvesicles ameliorate long-term bladder ischemia-induced bladder underactivity. J Formos Med Assoc 2024:S0929-6646(24)00565-5. [PMID: 39658414 DOI: 10.1016/j.jfma.2024.12.006] [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: 10/04/2024] [Revised: 11/01/2024] [Accepted: 12/03/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND/PURPOSE The mechanism for long-term hypoxia/ischemia induced bladder underactivity is uncertain. It requires an effectively therapeutic treatment. Therefore, we determined the pathophysiologic mechanisms of long-term bilateral partial iliac arterial occlusion (BPAO)-induced bladder underactivity and explored the therapeutic potential of adipose-derived stem cells (ADSCs) and ADSC-derived microvesicles (MVs) on BPAO-induced bladder dysfunction. METHODS The study included four groups: sham, BPAO, BPAO + ADSCs, and BPAO + ADSC-MVs. ADSCs or ADSC-MVs were isolated, characterized with specific CD markers and injected through the femoral artery to the rat bladders. Real-time laser speckle contrast imaging evaluated bladder microcirculation after BPAO. The transcystometrogram, pelvic nerve activity, bladder histology, immunohistochemistry, and lipid peroxidation assays were conducted after 4-week BPAO induction. The molecular mechanisms of bladder expression of purinergic P2X2/P2X3 and cholinergic M2/M3 receptors for regulating bladder contractility, nerve growth factor (NGF) for nerve injury repair, and collagen-1 for fibrosis were evaluated. RESULTS Long-term BPAO significantly reduced bladder microcirculation, prolonged the intercontraction interval, decreased voiding volume, increased residual urine volume, lengthened phase 1 contraction, shortened phase 2 contraction, increased leukocytes and CD68 infiltration, increased malondialdehyde levels, and decreased levels of P2X3 and M3 receptors. ADSC-MVs were more efficient than ADSCs in improving BPAO induced parameters, recovering P2X3 and M3 receptors, increasing NGF expression, and decreasing collagen-1 expression in the bladder. CONCLUSIONS ADSC-derived MVs were better than ADSCs to improve long-term BPAO-induced detrusor underactivity, bladder ischemia, and oxidative stress. ADSC-MVs through the therapeutic action of ameliorating inflammation, improving purinergic/cholinergic signaling and neuronal regeneration, and decreasing fibrosis improved BPAO-induced bladder underactivity.
Collapse
Affiliation(s)
- Bing-Juin Chiang
- College of Medicine, Fu-Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City, 24205, Taiwan; Department of Urology, Cardinal Tien Hospital, No.362, Zhongzheng Rd., Xindian Dist., New Taipei City, 23148, Taiwan; Department of Life Science, College of Science, National Taiwan Normal University, 162, Section 1, Heping E. Rd., Taipei, 106, Taiwan
| | - Su-Han Mao
- College of Medicine, Fu-Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City, 24205, Taiwan; Department of Urology, Cardinal Tien Hospital, No.362, Zhongzheng Rd., Xindian Dist., New Taipei City, 23148, Taiwan; Department of Life Science, College of Science, National Taiwan Normal University, 162, Section 1, Heping E. Rd., Taipei, 106, Taiwan
| | - Tung-Sheng Chen
- Department of Life Science, College of Science, National Taiwan Normal University, 162, Section 1, Heping E. Rd., Taipei, 106, Taiwan.
| | - Shiu-Dong Chung
- Division of Urology, Department of Surgery, Far-Eastern Memorial Hospital, No. 21, Section 2, Nanya S. Road, Banqiao Dist., New Taipei City, 220, Taiwan; Department of Nursing, College of Healthcare & Management, Asia Eastern University of Science and Technology, No.58, Sec.2, Sihchuan Rd., Banciao District, New Taipei City, 220303, Taiwan; General Education Center, Asia Eastern University of Science and Technology, No.58, Sec.2, Sihchuan Rd., Banciao District, New Taipei City, 220303, Taiwan.
| | - Chiang-Ting Chien
- Department of Life Science, College of Science, National Taiwan Normal University, 162, Section 1, Heping E. Rd., Taipei, 106, Taiwan.
| |
Collapse
|
4
|
Jafar H, Alqudah D, Rahmeh R, Al-Hattab D, Ahmed K, Rayyan R, Abusneinah A, Rasheed M, Rayyan Y, Awidi A. Safety and Potential Efficacy of Expanded Umbilical Cord-Derived Mesenchymal Stromal Cells in Luminal Ulcerative Colitis Patients. Stem Cells Dev 2024; 33:645-651. [PMID: 39446772 DOI: 10.1089/scd.2024.0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by periods of flare-ups and remission. It is likely to be an autoimmune in origin, presenting persistent therapeutic challenges despite current therapies. This study aims to investigate the potential of umbilical cord mesenchymal stromal cells (UCMSCs) in treating ulcerative colitis (UC). This study is a prospective phase 1 pilot, open-label, single-arm, and single-center study. UCMSCs were cultured under current Good Manufacturing Practice (cGMP) conditions and intravenously administered to six patients with UC. Safety and efficacy were evaluated using the Mayo Score/Disease Activity Index. Among the six enrolled adult patients, five completed long-term follow-ups. All exhibited at diagnosis active UC confirmed through comprehensive assessment methods. Each patient received three injections intravenously 2 weeks apart with a dose of 100 million UCMSC each. No significant short-term or intermediate-term adverse events were detected post-UCMSC administration. Long-term follow-up at 12 and 24 months showed sustained safety and no adverse events. Notably, three out of five patients achieved a Mayo score of 0 for UC, maintained at both 12 and 24 months, indicating a highly significant response (P < 0.001). This study demonstrates the safety and potential efficacy of UCMSCs in active UC. However, larger trials are warranted to validate these preliminary findings and to establish the role of UCMSC therapy as an option for managing UC.
Collapse
Affiliation(s)
- Hanan Jafar
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Dana Alqudah
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Reem Rahmeh
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Dana Al-Hattab
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Electrical and Mathematical Sciences and Engineering Department, King Abdulla University of Science and Technology, Thuwal, ThuwalSaudi Arabia
| | - Khalid Ahmed
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Rama Rayyan
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Awni Abusneinah
- School of Medicine, The University of Jordan, Amman, Jordan
- Internal Medicine Department, Jordan University Hospital, Amman, Jordan
| | - Mohammad Rasheed
- School of Medicine, The University of Jordan, Amman, Jordan
- Internal Medicine Department, Jordan University Hospital, Amman, Jordan
| | - Yaser Rayyan
- School of Medicine, The University of Jordan, Amman, Jordan
- Internal Medicine Department, Jordan University Hospital, Amman, Jordan
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- School of Medicine, The University of Jordan, Amman, Jordan
- Internal Medicine Department, Jordan University Hospital, Amman, Jordan
| |
Collapse
|
5
|
Al Monla R, Daien V, Michon F. Advanced bioengineering strategies broaden the therapeutic landscape for corneal failure. Front Bioeng Biotechnol 2024; 12:1480772. [PMID: 39605752 PMCID: PMC11598527 DOI: 10.3389/fbioe.2024.1480772] [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: 08/14/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
The cornea acts as the eye foremost protective layer and is essential for its focusing power. Corneal blindness may arise from physical trauma or conditions like dystrophies, keratitis, keratoconus, or ulceration. While conventional treatments involve medical therapies and donor allografts-sometimes supplemented with keratoprostheses-these options are not suitable for all corneal defects. Consequently, the development of bioartificial corneal tissue has emerged as a critical research area, aiming to address the global shortage of human cornea donors. Bioengineered corneas hold considerable promise as substitutes, with the potential to replace either specific layers or the entire thickness of damaged corneas. This review first delves into the structural anatomy of the human cornea, identifying key attributes necessary for successful corneal tissue bioengineering. It then examines various corneal pathologies, current treatments, and their limitations. Finally, the review outlines the primary approaches in corneal tissue engineering, exploring cell-free, cell-based, and scaffold-based options as three emerging strategies to address corneal failure.
Collapse
Affiliation(s)
- Reem Al Monla
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
| | - Vincent Daien
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
- Sydney Medical School, The Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Frederic Michon
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
| |
Collapse
|
6
|
Liu L, Fandiño J, McCarthy SD, Masterson CH, Sallent I, Du S, Warren A, Laffey JG, O’Toole D. The Effects of the Pneumonia Lung Microenvironment on MSC Function. Cells 2024; 13:1581. [PMID: 39329762 PMCID: PMC11430541 DOI: 10.3390/cells13181581] [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: 07/02/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Despite promise in preclinical models of acute respiratory distress syndrome (ARDS), mesenchymal stem cells (MSC) have failed to translate to therapeutic benefit in clinical trials. The MSC is a live cell medicine and interacts with the patient's disease state. Here, we explored this interaction, seeking to devise strategies to enhance MSC therapeutic function. METHODS Human bone-marrow-derived MSCs were exposed to lung homogenate from healthy and E. coli-induced ARDS rat models. Apoptosis and functional assays of the MSCs were performed. RESULTS The ARDS model showed reduced arterial oxygenation, decreased lung compliance and an inflammatory microenvironment compared to controls. MSCs underwent more apoptosis after stimulation by lung homogenate from controls compared to E. coli, which may explain why MSCs persist longer in ARDS subjects after administration. Changes in expression of cell surface markers and cytokines were associated with lung homogenate from different groups. The anti-microbial effects of MSCs did not change with the stimulation. Moreover, the conditioned media from lung-homogenate-stimulated MSCs inhibited T-cell proliferation. CONCLUSIONS These findings suggest that the ARDS microenvironment plays an important role in the MSC's therapeutic mechanism of action, and changes can inform strategies to modulate MSC-based cell therapy for ARDS.
Collapse
Affiliation(s)
- Lanzhi Liu
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- Discipline of Physiology, University of Galway, H91 W5P7 Galway, Ireland
| | - Juan Fandiño
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- School of Medicine, University of Galway, H91 W5P7 Galway, Ireland
| | - Sean D. McCarthy
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- School of Medicine, University of Galway, H91 W5P7 Galway, Ireland
| | - Claire H. Masterson
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- School of Medicine, University of Galway, H91 W5P7 Galway, Ireland
| | - Ignacio Sallent
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- School of Medicine, University of Galway, H91 W5P7 Galway, Ireland
| | - Shanshan Du
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- School of Medicine, University of Galway, H91 W5P7 Galway, Ireland
| | - Abigail Warren
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- Discipline of Anaesthesia, University of Galway, H91 V4AY Galway, Ireland
| | - John G. Laffey
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- Discipline of Anaesthesia, University of Galway, H91 V4AY Galway, Ireland
- Anaesthesia and Critical Care, Galway University Hospital, H91 V4AY Galway, Ireland
| | - Daniel O’Toole
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- Discipline of Physiology, University of Galway, H91 W5P7 Galway, Ireland
| |
Collapse
|
7
|
Pignatti E, Maccaferri M, Pisciotta A, Carnevale G, Salvarani C. A comprehensive review on the role of mesenchymal stromal/stem cells in the management of rheumatoid arthritis. Expert Rev Clin Immunol 2024; 20:463-484. [PMID: 38163928 DOI: 10.1080/1744666x.2023.2299729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease with systemic manifestations. Although the success of immune modulatory drug therapy is considerable, about 40% of patients do not respond to treatment. Mesenchymal stromal/stem cells (MSCs) have been demonstrated to have therapeutic potential for inflammatory diseases. AREAS COVERED This review provides an update on RA disease and on pre-clinical and clinical studies using MSCs from bone marrow, umbilical cord, adipose tissue, and dental pulp, to regulate the immune response. Moreover, the clinical use, safety, limitations, and future perspective of MSCs in RA are discussed. Using the PubMed database and ClincalTrials.gov, peer-reviewed full-text papers, abstracts and clinical trials were identified from 1985 through to April 2023. EXPERT OPINION MSCs demonstrated a satisfactory safety profile and potential for clinical efficacy. However, it is mandatory to deepen the investigations on how MSCs affect the proinflammatory deregulated RA patients' cells. MSCs are potentially good candidates for severe RA patients not responding to conventional therapies but a long-term follow-up after stem cells treatment and standardized protocols are needed. Future research should focus on well-designed multicenter randomized clinical trials with adequate sample sizes and properly selected patients satisfying RA criteria for a valid efficacy evaluation.
Collapse
Affiliation(s)
- Elisa Pignatti
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Monia Maccaferri
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Pisciotta
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Gianluca Carnevale
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Salvarani
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Rheumatology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
8
|
Gill JK, Rehsia SK, Verma E, Sareen N, Dhingra S. Stem cell therapy for cardiac regeneration: past, present, and future. Can J Physiol Pharmacol 2024; 102:161-179. [PMID: 38226807 DOI: 10.1139/cjpp-2023-0202] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Cardiac disorders remain the leading cause of mortality worldwide. Current clinical strategies, including drug therapy, surgical interventions, and organ transplantation offer limited benefits to patients without regenerating the damaged myocardium. Over the past decade, stem cell therapy has generated a keen interest owing to its unique self-renewal and immune privileged characteristics. Furthermore, the ability of stem cells to differentiate into specialized cell types, has made them a popular therapeutic tool against various diseases. This comprehensive review provides an overview of therapeutic potential of different types of stem cells in reference to cardiovascular diseases. Furthermore, it sheds light on the advantages and limitations associated with each cell type. An in-depth analysis of the challenges associated with stem cell research and the hurdles for its clinical translation and their possible solutions have also been elaborated upon. It examines the controversies surrounding embryonic stem cells and the emergence of alternative approaches, such as the use of induced pluripotent stem cells for cardiac therapeutic applications. Overall, this review serves as a valuable resource for researchers, clinicians, and policymakers involved in the field of regenerative medicine, guiding the development of safe and effective stem cell-based therapies to revolutionize patient care.
Collapse
Affiliation(s)
- Jaideep Kaur Gill
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Biomedical Engineering Program, University of Manitoba, Winnipeg MB, R2H2A6, Canada
| | - Sargun Kaur Rehsia
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Biomedical Engineering Program, University of Manitoba, Winnipeg MB, R2H2A6, Canada
| | - Elika Verma
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Biomedical Engineering Program, University of Manitoba, Winnipeg MB, R2H2A6, Canada
| | - Niketa Sareen
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Biomedical Engineering Program, University of Manitoba, Winnipeg MB, R2H2A6, Canada
| | - Sanjiv Dhingra
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Biomedical Engineering Program, University of Manitoba, Winnipeg MB, R2H2A6, Canada
| |
Collapse
|
9
|
Petrella F, Cassina EM, Libretti L, Pirondini E, Raveglia F, Tuoro A. Mesenchymal Stromal Cell Therapy for Thoracic Surgeons: An Update. J Pers Med 2023; 13:1632. [PMID: 38138859 PMCID: PMC10744666 DOI: 10.3390/jpm13121632] [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: 10/20/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Stem cells are undifferentiated cells presenting extensive self-renewal features and the ability to differentiate "in vitro" and "in vivo" into a range of lineage cells, like chondrogenic, osteogenic and adipogenic lineages when cultured in specific inducing media. Two major domains of clinical applications of stem cells in thoracic surgery have been investigated: regenerative medicine, which is a section of translational research in tissue engineering focusing on the replacement, renewal or regeneration of cells, tissues and organs to re-establish damaged physiologic functions; drug loading and delivery, representing a new branch proposing stem cells as carriers to provide selected districts with anti-cancer agents for targeted treatments.
Collapse
Affiliation(s)
- Francesco Petrella
- Department of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (E.M.C.); (L.L.); (E.P.); (F.R.); (A.T.)
| | | | | | | | | | | |
Collapse
|
10
|
Arango-Rodríguez ML, Mateus LC, Sossa CL, Becerra-Bayona SM, Solarte-David VA, Ochoa Vera ME, Viviescas LTG, Berrio AMV, Serrano SE, Vargas O, Isla AC, Benitez A, Rangel G. A novel therapeutic management for diabetes patients with chronic limb-threatening ischemia: comparison of autologous bone marrow mononuclear cells versus allogenic Wharton jelly-derived mesenchymal stem cells. Stem Cell Res Ther 2023; 14:221. [PMID: 37626416 PMCID: PMC10464344 DOI: 10.1186/s13287-023-03427-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Chronic limb-threatening ischemia (CLTI) represents the final stage of peripheral arterial disease. Approximately one-third of patients with CLTI are not eligible for conventional surgical treatments. Furthermore, patients with advanced stage of CLTI are prone to amputation and death. Thus, an effective therapeutic strategy is urgently needed. In this context, autologous bone marrow mononuclear cell (auto-BM-MNC) and allogeneic mesenchymal stem cells represent a promising therapeutic approach for treating CLTI. In this study, we compared the safety and beneficial therapeutic effect of auto-BM-MNC versus allogeneic Wharton jelly-derived mesenchymal stem cells (allo-WJ-MSCs) in diabetic patients with CLTI. METHODS We performed a randomized, prospective, double-blind and controlled pilot study. Twenty-four diabetic patients in the advanced stage of CLTI (4 or 5 in Rutherford's classification) and a transcutaneous oxygen pressure (TcPO2) below 30 mmHg were randomized to receive 15 injections of (i) auto-BM-MNC (7.197 × 106 ± 2.984 × 106 cells/mL) (n = 7), (ii) allo-WJ-MSCs (1.333 × 106 cells/mL) (n = 7) or (iii) placebo solution (1 mL) (n = 10), which were administered into the periadventitial layer of the arterial walls under eco-Doppler guidance. The follow-up visits were at months 1, 3, 6, and 12 to evaluate the following parameters: (i) Rutherford's classification, (ii) TcPO2, (iii) percentage of wound closure, (iv) pain, (v) pain-free walking distance, (vi) revascularization and limb-survival proportion, and (vii) life quality (EQ-5D questionnaire). RESULTS No adverse events were reported. Patients with CLTI who received auto-BM-MNC and allo-WJ-MSCs presented an improvement in Rutherford's classification, a significant increase in TcPO2 values, a reduction in the lesion size in a shorter time, a decrease in the pain score and an increase in the pain-free walking distance, in comparison with the placebo group. In addition, the participants treated with auto-BM-MNC and allo-WJ-MSCs kept their limbs during the follow-up period, unlike the placebo group, which had a marked increase in amputation. CONCLUSIONS Our results showed that patients with CLTI treated with auto-BM-MNC and allo-WJ-MSCs conserved 100% of their limb during 12 months of the follow-up compared to the placebo group, where 60% of participants underwent limb amputation in different times. Furthermore, we observed a faster improvement in the allo-WJ-MSC group, unlike the auto-BM-MNC group. Trial registration This study was retrospectively registered at ClinicalTrials.gov (NCT05631444).
Collapse
Affiliation(s)
- Martha L Arango-Rodríguez
- Banco Multitejidos y Centro de Terapias Avanzadas, Clínica FOSCAL Internacional, 681004, Floridablanca, Colombia.
| | - Ligia C Mateus
- Fundación Oftalmológica de Santander Carlos Ardila Lulle, 681004, Floridablanca, Colombia
| | - Claudia L Sossa
- Fundación Oftalmológica de Santander Carlos Ardila Lulle, 681004, Floridablanca, Colombia
- Programa para el Tratamiento y Estudio de Enfermedades Hematológicas y Oncológicas de Santander (PROTEHOS), 681004153, Floridablanca, Colombia
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, 681003, Bucaramanga, Colombia
| | - Silvia M Becerra-Bayona
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, 681003, Bucaramanga, Colombia
| | - Víctor Alfonso Solarte-David
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, 681003, Bucaramanga, Colombia
- Facultad de Ingeniería, Universidad Autónoma de Bucaramanga - UNAB, 680003, Bucaramanga, Colombia
| | - Miguel Enrique Ochoa Vera
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, 681003, Bucaramanga, Colombia
| | - Lady T Giratá Viviescas
- Banco Multitejidos y Centro de Terapias Avanzadas, Clínica FOSCAL Internacional, 681004, Floridablanca, Colombia
| | - Ana M Vera Berrio
- Banco Multitejidos y Centro de Terapias Avanzadas, Clínica FOSCAL Internacional, 681004, Floridablanca, Colombia
| | - Sergio Eduardo Serrano
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, 681003, Bucaramanga, Colombia
| | - Oliverio Vargas
- Fundación Oftalmológica de Santander Carlos Ardila Lulle, 681004, Floridablanca, Colombia
| | - Andrés Catalá Isla
- Fundación Oftalmológica de Santander Carlos Ardila Lulle, 681004, Floridablanca, Colombia
| | - Alape Benitez
- Fundación Oftalmológica de Santander Carlos Ardila Lulle, 681004, Floridablanca, Colombia
| | - Germán Rangel
- Fundación Oftalmológica de Santander Carlos Ardila Lulle, 681004, Floridablanca, Colombia
| |
Collapse
|
11
|
Clavellina D, Balkan W, Hare JM. Stem cell therapy for acute myocardial infarction: Mesenchymal Stem Cells and induced Pluripotent Stem Cells. Expert Opin Biol Ther 2023; 23:951-967. [PMID: 37542462 PMCID: PMC10837765 DOI: 10.1080/14712598.2023.2245329] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/07/2023]
Abstract
INTRODUCTION Acute myocardial infarction (AMI) remains a leading cause of death in the United States. The limited capacity of cardiomyocytes to regenerate and the restricted contractility of scar tissue after AMI are not addressed by current pharmacologic interventions. Mesenchymal stem/stromal cells (MSCs) have emerged as a promising therapeutic approach due to their low antigenicity, ease of harvesting, and efficacy and safety in preclinical and clinical studies, despite their low survival and engraftment rates. Other stem cell types, such as induced pluripotent stem cells (iPSCs) also show promise, and optimizing cardiac repair requires integrating emerging technologies and strategies. AREAS COVERED This review offers insights into advancing cell-based therapies for AMI, emphasizing meticulously planned trials with a standardized definition of AMI, for a bench-to-bedside approach. We critically evaluate fundamental studies and clinical trials to provide a comprehensive overview of the advances, limitations and prospects for cell-based therapy in AMI. EXPERT OPINION MSCs continue to show potential promise for treating AMI and its sequelae, but addressing their low survival and engraftment rates is crucial for clinical success. Integrating emerging technologies such as pluripotent stem cells and conducting well-designed trials will harness the full potential of cell-based therapy in AMI management. Collaborative efforts are vital to developing effective stem cell therapies for AMI patients.
Collapse
Affiliation(s)
- Diana Clavellina
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
12
|
Mattoli S, Schmidt M. Investigational Use of Mesenchymal Stem/Stromal Cells and Their Secretome as Add-On Therapy in Severe Respiratory Virus Infections: Challenges and Perspectives. Adv Ther 2023; 40:2626-2692. [PMID: 37069355 PMCID: PMC10109238 DOI: 10.1007/s12325-023-02507-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/24/2023] [Indexed: 04/19/2023]
Abstract
Serious manifestations of respiratory virus infections such as influenza and coronavirus disease 2019 (COVID-19) are associated with a dysregulated immune response and systemic inflammation. Treating the immunological/inflammatory dysfunction with glucocorticoids, Janus kinase inhibitors, and monoclonal antibodies against the interleukin-6 receptor has significantly reduced the risk of respiratory failure and death in hospitalized patients with severe COVID-19, but the proportion of those requiring invasive mechanical ventilation (IMV) and dying because of respiratory failure remains elevated. Treatment of severe influenza-associated pneumonia and acute respiratory distress syndrome (ARDS) with available immunomodulators and anti-inflammatory compounds is still not recommended. New therapies are therefore needed to reduce the use of IMV and the risk of death in hospitalized patients with rapidly increasing oxygen demand and systemic inflammation who do not respond to the current standard of care. This paper provides a critical assessment of the published clinical trials that have tested the investigational use of intravenously administered allogeneic mesenchymal stem/stromal cells (MSCs) and MSC-derived secretome with putative immunomodulatory/antiinflammatory/regenerative properties as add-on therapy to improve the outcome of these patients. Increased survival rates are reported in 5 of 12 placebo-controlled or open-label comparative trials involving patients with severe and critical COVID-19 and in the only study concerning patients with influenza-associated ARDS. Results are encouraging but inconclusive for the following reasons: small number of patients tested in each trial; differences in concomitant treatments and respiratory support; imbalances between study arms; differences in MSC source, MSC-derived product, dosing and starting time of the investigational therapy; insufficient/inappropriate reporting of clinical data. Solutions are proposed for improving the clinical development plan, with the aim of facilitating regulatory approval of the MSC-based investigational therapy for life-threatening respiratory virus infections in the future. Major issues are the absence of a biomarker predicting responsiveness to MSCs and MSC-derived secretome and the lack of pharmacoeconomic evaluations.
Collapse
Affiliation(s)
- Sabrina Mattoli
- Center of Expertise in Research and Innovation of the International Network for the Advancement of Viable and Applicable Innovations in Life Sciences (InAvail), InAvail at Rosental Nexxt, 4058 Basel, Switzerland
- Avail Biomedical Research Institute, 80539 Munich, Germany
| | - Matthias Schmidt
- Avail Biomedical Research Institute, 80539 Munich, Germany
- Discovery and Translational Research Center, 80539 Munich, Germany
| |
Collapse
|
13
|
Byrnes D, Masterson CH, Gonzales HE, McCarthy SD, O’Toole DP, Laffey JG. Multiple Dosing and Preactivation of Mesenchymal Stromal Cells Enhance Efficacy in Established Pneumonia Induced by Antimicrobial-Resistant Klebsiella pneumoniae in Rodents. Int J Mol Sci 2023; 24:8055. [PMID: 37175761 PMCID: PMC10179238 DOI: 10.3390/ijms24098055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Antimicrobial-resistant (AMR) bacteria, such as Klebsiella species, are an increasingly common cause of hospital-acquired pneumonia, resulting in high mortality and morbidity. Harnessing the host immune response to AMR bacterial infection using mesenchymal stem cells (MSCs) is a promising approach to bypass bacterial AMR mechanisms. The administration of single doses of naïve MSCs to ARDS clinical trial patient cohorts has been shown to be safe, although efficacy is unclear. The study tested whether repeated MSC dosing and/or preactivation, would attenuate AMR Klebsiella pneumonia-induced established pneumonia. Rat models of established K. pneumoniae-induced pneumonia were randomised to receive intravenous naïve or cytomix-preactivated umbilical cord MSCs as a single dose at 24 h post pneumonia induction with or without a subsequent dose at 48 h. Physiological indices, bronchoalveolar lavage (BAL), and tissues were obtained at 72 h post pneumonia induction. A single dose of naïve MSCs was largely ineffective, whereas two doses of MSCs were effective in attenuating Klebsiella pneumosepsis, improving lung compliance and oxygenation, while reducing bacteria and injury in the lung. Cytomix-preactivated MSCs were superior to naïve MSCs. BAL neutrophil counts and activation were reduced, and apoptosis increased. MSC therapy reduced cytotoxic BAL T cells, and increased CD4+/CD8+ ratios. Systemically, granulocytes, classical monocytes, and the CD4+/CD8+ ratio were reduced, and nonclassical monocytes were increased. Repeated doses of MSCs-particularly preactivated MSCs-enhance their therapeutic potential in a clinically relevant model of established AMR K. pneumoniae-induced pneumosepsis.
Collapse
Affiliation(s)
- Declan Byrnes
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
| | - Claire H. Masterson
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
| | - Hector E. Gonzales
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
| | - Sean D. McCarthy
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
| | - Daniel P. O’Toole
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
| | - John G. Laffey
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
- Department of Anaesthesia, Galway University Hospitals, SAOLTA University Health Group, H91 YR71 Galway, Ireland
| |
Collapse
|
14
|
Knudsen L, Hummel B, Wrede C, Zimmermann R, Perlman CE, Smith BJ. Acinar micromechanics in health and lung injury: what we have learned from quantitative morphology. Front Physiol 2023; 14:1142221. [PMID: 37025383 PMCID: PMC10070844 DOI: 10.3389/fphys.2023.1142221] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Within the pulmonary acini ventilation and blood perfusion are brought together on a huge surface area separated by a very thin blood-gas barrier of tissue components to allow efficient gas exchange. During ventilation pulmonary acini are cyclically subjected to deformations which become manifest in changes of the dimensions of both alveolar and ductal airspaces as well as the interalveolar septa, composed of a dense capillary network and the delicate tissue layer forming the blood-gas barrier. These ventilation-related changes are referred to as micromechanics. In lung diseases, abnormalities in acinar micromechanics can be linked with injurious stresses and strains acting on the blood-gas barrier. The mechanisms by which interalveolar septa and the blood-gas barrier adapt to an increase in alveolar volume have been suggested to include unfolding, stretching, or changes in shape other than stretching and unfolding. Folding results in the formation of pleats in which alveolar epithelium is not exposed to air and parts of the blood-gas barrier are folded on each other. The opening of a collapsed alveolus (recruitment) can be considered as an extreme variant of septal wall unfolding. Alveolar recruitment can be detected with imaging techniques which achieve light microscopic resolution. Unfolding of pleats and stretching of the blood-gas barrier, however, require electron microscopic resolution to identify the basement membrane. While stretching results in an increase of the area of the basement membrane, unfolding of pleats and shape changes do not. Real time visualization of these processes, however, is currently not possible. In this review we provide an overview of septal wall micromechanics with focus on unfolding/folding as well as stretching. At the same time we provide a state-of-the-art design-based stereology methodology to quantify microarchitecture of alveoli and interalveolar septa based on different imaging techniques and design-based stereology.
Collapse
Affiliation(s)
- Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Benjamin Hummel
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Christoph Wrede
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Richard Zimmermann
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Carrie E Perlman
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, United States
| | - Bradford J Smith
- Department of Bioengineering, College of Engineering Design and Computing, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, United States
- Department of Pediatric Pulmonary and Sleep Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
15
|
Hudock MR, Pinezich MR, Mir M, Chen J, Bacchetta M, Vunjak-Novakovic G, Kim J. Emerging Imaging Modalities for Functional Assessment of Donor Lungs Ex Vivo. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2023; 25:100432. [PMID: 36778755 PMCID: PMC9913406 DOI: 10.1016/j.cobme.2022.100432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The severe shortage of functional donor lungs that can be offered to recipients has been a major challenge in lung transplantation. Innovative ex vivo lung perfusion (EVLP) and tissue engineering methodologies are now being developed to repair damaged donor lungs that are deemed unsuitable for transplantation. To assess the efficacy of donor lung reconditioning methods intended to rehabilitate rejected donor lungs, monitoring of lung function with improved spatiotemporal resolution is needed. Recent developments in live imaging are enabling non-destructive, direct, and longitudinal modalities for assessing local tissue and whole lung functions. In this review, we describe how emerging live imaging modalities can be coupled with lung tissue engineering approaches to promote functional recovery of ex vivo donor lungs.
Collapse
Affiliation(s)
- Maria R. Hudock
- Department of Biomedical Engineering, Columbia University,
New York, NY, USA
| | - Meghan R. Pinezich
- Department of Biomedical Engineering, Columbia University,
New York, NY, USA
| | - Mohammad Mir
- Department of Biomedical Engineering, Stevens Institute of
Technology, Hoboken, NJ, USA
| | - Jiawen Chen
- Department of Biomedical Engineering, Stevens Institute of
Technology, Hoboken, NJ, USA
| | - Matthew Bacchetta
- Department of Cardiac Surgery, Vanderbilt University,
Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt
University, Nashville, TN, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University,
New York, NY, USA
- Department of Medicine, Columbia University, New York, NY,
USA
| | - Jinho Kim
- Department of Biomedical Engineering, Stevens Institute of
Technology, Hoboken, NJ, USA
| |
Collapse
|
16
|
Dos Santos CC, Vaswani CM, Mei SHJ, Rocco PRM, Weiss DJ, Stewart DJ, Liles WC. Reply: Mesenchymal stromal (stem) cell therapy modulates miR-193b-5p expression to attenuate sepsis-induced acute lung injury. Eur Respir J 2023; 61:13993003.00886-2022. [PMID: 36758998 DOI: 10.1183/13993003.00886-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 12/29/2022] [Indexed: 02/11/2023]
Affiliation(s)
- Claudia C Dos Santos
- The Keenan Research Centre for Biomedical Science of St Michael's Hospital, Toronto, ON, Canada
- Institute of Medical Sciences and Interdepartmental Division of Critical Care, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Chirag M Vaswani
- The Keenan Research Centre for Biomedical Science of St Michael's Hospital, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shirley H J Mei
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Duncan J Stewart
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - W Conrad Liles
- Department of Medicine and Sepsis Center of Research Excellence-UW (SCORE-UW), University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
Differential Effects of Cytokine Versus Hypoxic Preconditioning of Human Mesenchymal Stromal Cells in Pulmonary Sepsis Induced by Antimicrobial-Resistant Klebsiella pneumoniae. Pharmaceuticals (Basel) 2023. [DOI: 10.3390/ph16020149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background: Pulmonary sepsis is a leading cause of hospital mortality, and sepses arising from antimicrobial-resistant (AMR) bacterial strains are particularly difficult to treat. Here we investigated the potential of mesenchymal stromal cells (MSCs) to combat established Klebsiella pneumoniae pneumosepsis and further evaluated MSC preconditioning and pre-activation methods. Methods: The potential for naïve and preconditioned MSCs to enhance wound healing, reduce inflammation, preserve metabolic activity, and enhance bacterial killing was assessed in vitro. Rats were subjected to intratracheal K. pneumoniae followed by the intravenous administration of MSCs. Physiological indices, blood, bronchoalveolar lavage (BAL), and tissues were obtained 72 h later. Results: In vitro assays confirmed that preconditioning enhances MSC function, accelerating pulmonary epithelial wound closure, reducing inflammation, attenuating cell death, and increasing bacterial killing. Cytomix-pre-activated MSCs are superior to naïve and hypoxia-exposed MSCs in attenuating Klebsiella pneumosepsis, improving lung compliance and oxygenation, reducing bacteria, and attenuating histologic injuries in lungs. BAL inflammatory cytokines were reduced, correlating with decreases in polymorphonuclear (PMN) cells. MSCs increased PMN apoptosis and the CD4:CD8 ratio in BAL. Systemically, granulocytes, classical monocytes, and the CD4:CD8 ratio were reduced, and nonclassical monocytes were increased. Conclusions: Preconditioning with cytokines, but not hypoxia, enhances the therapeutic potential of MSCs in clinically relevant models of K. pneumoniae-induced pneumosepsis.
Collapse
|
18
|
Saccu G, Menchise V, Gai C, Bertolin M, Ferrari S, Giordano C, Manco M, Dastrù W, Tolosano E, Bussolati B, Calautti E, Camussi G, Altruda F, Fagoonee S. Bone Marrow Mesenchymal Stromal/Stem Cell-Derived Extracellular Vesicles Promote Corneal Wound Repair by Regulating Inflammation and Angiogenesis. Cells 2022; 11:3892. [PMID: 36497151 PMCID: PMC9736484 DOI: 10.3390/cells11233892] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Severe corneal damage leads to complete vision loss, thereby affecting life quality and impinging heavily on the healthcare system. Current clinical approaches to manage corneal wounds suffer from severe drawbacks, thus requiring the development of alternative strategies. Of late, mesenchymal stromal/stem cell (MSC)-derived extracellular vesicles (EVs) have become a promising tool in the ophthalmic field. In the present study, we topically delivered bone-marrow-derived MSC-EVs (BMSC-EVs), embedded in methylcellulose, in a murine model of alkali-burn-induced corneal damage in order to evaluate their role in corneal repair through histological and molecular analyses, with the support of magnetic resonance imaging. Our data show that BMSC-EVs, used for the first time in this specific formulation on the damaged cornea, modulate cell death, inflammation and angiogenetic programs in the injured tissue, thus leading to a faster recovery of corneal damage. These results were confirmed on cadaveric donor-derived human corneal epithelial cells in vitro. Thus, BMSC-EVs modulate corneal repair dynamics and are promising as a new cell-free approach for intervening on burn wounds, especially in the avascularized region of the eye.
Collapse
Affiliation(s)
- Gabriele Saccu
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Valeria Menchise
- Institute of Biostructure and Bioimaging, National Research Council (CNR), Molecular Biotechnology Center “Guido Tarone”, 10126 Turin, Italy
| | - Chiara Gai
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | | | | | - Cristina Giordano
- Ophthalmology Veterinary Practice, C.so Galileo Ferraris 121, 10126 Turin, Italy
| | - Marta Manco
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Walter Dastrù
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Emanuela Tolosano
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Benedetta Bussolati
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Enzo Calautti
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Fiorella Altruda
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging, National Research Council (CNR), Molecular Biotechnology Center “Guido Tarone”, 10126 Turin, Italy
| |
Collapse
|
19
|
Wang H, Jiang C, Cai J, Lu Q, Qiu Y, Wang Y, Huang Y, Xiao Y, Wang B, Wei X, Shi J, Lai X, Wang T, Wang J, Xiang AP. Nestin prevents mesenchymal stromal cells from apoptosis in LPS-induced lung injury via inhibition of unfolded protein response sensor IRE1α. LIFE MEDICINE 2022; 1:359-371. [PMID: 39872742 PMCID: PMC11749126 DOI: 10.1093/lifemedi/lnac049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/01/2022] [Indexed: 01/30/2025]
Abstract
The clinical applications of MSC therapy have been intensely investigated in acute respiratory distress syndrome. However, clinical studies have fallen short of expectations despite encouraging preclinical results. One of the key problems is that transplanted stem cells can hardly survive in the harsh inflammatory environment. Prolonging the survival of transplanted MSCs might be a promising strategy to enhance the therapeutic efficacy of MSC therapy. Here, we identified Nestin, a class VI intermediate filament, as a positive regulator of MSC survival in the inflammatory microenvironment. We showed that Nestin knockout led to a significant increase of MSC apoptosis, which hampered the therapeutic effects in an LPS-induced lung injury model. Mechanistically, Nestin knockout induced a significant elevation of endoplasmic reticulum (ER) stress level. Further investigations showed that Nestin could bind to IRE1α and inhibit ER stress-induced apoptosis under stress. Furthermore, pretreatment with IRE1α inhibitor 4μ8C improved MSC survival and improved therapeutic effect. Our data suggests that Nestin enhances stem cell survival after transplantation by inhibiting ER stress-induced apoptosis, improving protection, and repair of the lung inflammatory injury.
Collapse
Affiliation(s)
- Hongmiao Wang
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Chenhao Jiang
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Jianye Cai
- Department of Hepatic Surgery and Liver Transplantation Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
- Guangdong Key Laboratory of Liver Disease Research, Guangdong Engineering Laboratory for Transplantation, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Qiying Lu
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuan Qiu
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Yi Wang
- Guangdong Institute for Drug Control, NMPA Key Laboratory for Quality Control of Blood Products, Guangdong Drug Administration Key Laboratory of Quality Control and Research of Blood Products, Guangzhou 510663, China
| | - Yinong Huang
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Yong Xiao
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Boyan Wang
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoyue Wei
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiahao Shi
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Xingqiang Lai
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Tao Wang
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiancheng Wang
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
- Scientific Research Centre, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Andy Peng Xiang
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
20
|
Payushina OV, Tsomartova DA, Chereshneva YV, Ivanova MY, Lomanovskaya TA, Pavlova MS, Kuznetsov SL. Experimental Transplantation of Mesenchymal Stromal Cells as an Approach to Studying Their Differentiation In Vivo (Review). BIOL BULL+ 2022. [DOI: 10.1134/s1062359022060127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
21
|
Pichardo AH, Amadeo F, Wilm B, Lévy R, Ressel L, Murray P, Sée V. Optical Tissue Clearing to Study the Intra-Pulmonary Biodistribution of Intravenously Delivered Mesenchymal Stromal Cells and Their Interactions with Host Lung Cells. Int J Mol Sci 2022; 23:14171. [PMID: 36430651 PMCID: PMC9699424 DOI: 10.3390/ijms232214171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) injected intravenously are trapped in the capillaries of the lungs and die within the first 24 h. Studying the biodistribution and fate of labelled therapeutic cells in the 3D pulmonary context is important to understand their function in this organ and gain insights into their mechanisms of action. Optical tissue clearing enables volumetric cell tracking at single-cell resolution. Thus, we compared three optical tissue-clearing protocols (Clear, Unobstructed Brain/Body Imaging Cocktails and Computational analysis (CUBIC), modified stabilised 3D imaging of solvent-cleared organs (s-DISCO) and ethyl cinnamate (ECi)) to evaluate their potential to track the biodistribution of human umbilical cord MSCs expressing the tdTomato fluorescence reporter and investigate how they interact with host cells in the mouse lung. The results showed that although CUBIC clearing is the only method that enables direct imaging of fluorescently labelled MSCs, combining s-DISCO or ECi with immunofluorescence or dye labelling allows the interaction of MSCs with endothelial and immune cells to be studied. Overall, this comparative study offers guidance on selecting an optical tissue-clearing method for cell tracking applications.
Collapse
Affiliation(s)
- Alejandra Hernandez Pichardo
- Department of Molecular Physiology and Cell Signalling, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3BX, UK
- Centre for Preclinical Imaging, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Francesco Amadeo
- Department of Molecular Physiology and Cell Signalling, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3BX, UK
- Centre for Preclinical Imaging, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Bettina Wilm
- Department of Molecular Physiology and Cell Signalling, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3BX, UK
- Centre for Preclinical Imaging, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Raphaël Lévy
- INSERM, LVTS, Université Sorbonne Paris Nord, F-75018 Paris, France
| | - Lorenzo Ressel
- Department of Veterinary Anatomy Physiology and Pathology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Patricia Murray
- Department of Molecular Physiology and Cell Signalling, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3BX, UK
- Centre for Preclinical Imaging, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Violaine Sée
- CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), University Claude Bernard Lyon1, 69007 Lyon, France
| |
Collapse
|
22
|
Weiss DJ, Rolandsson Enes S. MSCs interaction with the host lung microenvironment: An overlooked aspect? Front Immunol 2022; 13:1072257. [DOI: 10.3389/fimmu.2022.1072257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) were identified more than 50 years ago, and research advances have promoted the translation of pre-clinical studies into clinical settings in several diseases. However, we are only starting to uncover the local factors that regulate cell phenotype, cell function, and cell viability across tissues following administration in different diseases. Advances in pre-clinical and translational studies suggest that the host environment, especially inflammatory active environments, plays a significant role in directing the infused MSCs towards different phenotypes with different functions. This can significantly effect their therapeutic efficacy. One way to study this interaction between the host environment and the infused cells is to expose MSCs ex vivo to patient samples such as serum or bronchoalveolar lavage fluid. Using this approach, it has been demonstrated that MSCs are very sensitive to different host factors such as pathogens, inflammatory cytokines, and extra cellular matrix properties. By understanding how different local host factors effect MSC function it will open possibilities to select specific patient sub-groups that are more likely to respond to this type of treatment and will also open possibilities to prime the local host environment to increase viability and to enrich for a specific MSC phenotype. Here, we aim to review the current understanding of the interaction of MSCs with the host microenvironment. To narrow the scope of this mini review, the focus will be on the pulmonary microenvironment, with a specific focus on the diseases acute respiratory distress syndrome (ARDS) and cystic fibrosis (CF).
Collapse
|
23
|
Tynecka M, Janucik A, Niemira M, Zbikowski A, Stocker N, Tarasik A, Starosz A, Grubczak K, Szalkowska A, Korotko U, Reszec J, Kwasniewski M, Kretowski A, Akdis C, Sokolowska M, Moniuszko M, Eljaszewicz A. The short-term and long-term effects of intranasal mesenchymal stem cell administration to noninflamed mice lung. Front Immunol 2022; 13:967487. [PMID: 36189248 PMCID: PMC9523259 DOI: 10.3389/fimmu.2022.967487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
Mesenchymal stem cells (mesenchymal stromal cells; MSC)-based therapies remain a promising approach to treat degenerative and inflammatory diseases. Their beneficial effects were confirmed in numerous experimental models and clinical trials. However, safety issues concerning MSCs’ stability and their long-term effects limit their implementation in clinical practice, including treatment of respiratory diseases such as asthma, chronic obstructive pulmonary disease, and COVID-19. Here, we aimed to investigate the safety of intranasal application of human adipose tissue-derived MSCs in a preclinical experimental mice model and elucidate their effects on the lungs. We assessed short-term (two days) and long-term (nine days) effects of MSCs administration on lung morphology, immune responses, epithelial barrier function, and transcriptomic profiles. We observed an increased frequency of IFNγ- producing T cells and a decrease in occludin and claudin 3 as a long-term effect of MSCs administration. We also found changes in the lung transcriptomic profiles, reflecting redox imbalance and hypoxia signaling pathway. Additionally, we found dysregulation in genes clustered in pattern recognition receptors, macrophage activation, oxidative stress, and phagocytosis. Our results suggest that i.n. MSCs administration to noninflamed healthy lungs induces, in the late stages, low-grade inflammatory responses aiming at the clearance of MSCs graft.
Collapse
Affiliation(s)
- Marlena Tynecka
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Janucik
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
| | - Magdalena Niemira
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Arkadiusz Zbikowski
- Department of Medical Biology, Medical University of Bialystok, Bialystok, Poland
| | - Nino Stocker
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | - Agnieszka Tarasik
- Department of Medical Pathomorphology, Medical University of Bialystok, Bialystok, Poland
| | - Aleksandra Starosz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
| | - Kamil Grubczak
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
| | - Anna Szalkowska
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Urszula Korotko
- Centre for Bioinformatics and Data Analysis, Medical University of Bialystok, Bialystok, Poland
| | - Joanna Reszec
- Department of Medical Pathomorphology, Medical University of Bialystok, Bialystok, Poland
| | - Miroslaw Kwasniewski
- Centre for Bioinformatics and Data Analysis, Medical University of Bialystok, Bialystok, Poland
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Cezmi Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
- Department of Allergology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
- *Correspondence: Andrzej Eljaszewicz, ; Marcin Moniuszko,
| | - Andrzej Eljaszewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
- *Correspondence: Andrzej Eljaszewicz, ; Marcin Moniuszko,
| |
Collapse
|
24
|
Yang D, Ortinau L, Jeong Y, Park D. Advances and challenges in intravital imaging of craniofacial and dental progenitor cells. Genesis 2022; 60:e23498. [PMID: 35980285 PMCID: PMC10015615 DOI: 10.1002/dvg.23498] [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: 03/30/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 11/11/2022]
Abstract
Craniofacial and appendicular bone homeostasis is dynamically regulated by a balance between bone formation and resorption by osteoblasts and osteoclasts, respectively. Despite the developments in multiple imaging techniques in bone biology, there are still technical challenges and limitations in the investigation of spatial/anatomical location of rare stem/progenitor cells and their molecular regulation in tooth and craniofacial bones of living animals. Recent advances in live animal imaging techniques for the craniofacial and dental apparatus can provide new insights in real time into bone stem/progenitor cell dynamics and function in vivo. Here, we review the current inventions and applications of the noninvasive intravital imaging technique and its practical uses and limitations in the analysis of stem/progenitor cells in craniofacial and dental apparatus in vivo. Furthermore, we also explore the potential applications of intravital microscopy in the dental field.
Collapse
Affiliation(s)
- Dongwook Yang
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Center for Skeletal Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Laura Ortinau
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Center for Skeletal Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Youngjae Jeong
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Center for Skeletal Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Dongsu Park
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Center for Skeletal Biology, Baylor College of Medicine, Houston, Texas, USA.,Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
25
|
Watson-Levings RS, Palmer GD, Levings PP, Dacanay EA, Evans CH, Ghivizzani SC. Gene Therapy in Orthopaedics: Progress and Challenges in Pre-Clinical Development and Translation. Front Bioeng Biotechnol 2022; 10:901317. [PMID: 35837555 PMCID: PMC9274665 DOI: 10.3389/fbioe.2022.901317] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
In orthopaedics, gene-based treatment approaches are being investigated for an array of common -yet medically challenging- pathologic conditions of the skeletal connective tissues and structures (bone, cartilage, ligament, tendon, joints, intervertebral discs etc.). As the skeletal system protects the vital organs and provides weight-bearing structural support, the various tissues are principally composed of dense extracellular matrix (ECM), often with minimal cellularity and vasculature. Due to their functional roles, composition, and distribution throughout the body the skeletal tissues are prone to traumatic injury, and/or structural failure from chronic inflammation and matrix degradation. Due to a mixture of environment and endogenous factors repair processes are often slow and fail to restore the native quality of the ECM and its function. In other cases, large-scale lesions from severe trauma or tumor surgery, exceed the body’s healing and regenerative capacity. Although a wide range of exogenous gene products (proteins and RNAs) have the potential to enhance tissue repair/regeneration and inhibit degenerative disease their clinical use is hindered by the absence of practical methods for safe, effective delivery. Cumulatively, a large body of evidence demonstrates the capacity to transfer coding sequences for biologic agents to cells in the skeletal tissues to achieve prolonged delivery at functional levels to augment local repair or inhibit pathologic processes. With an eye toward clinical translation, we discuss the research progress in the primary injury and disease targets in orthopaedic gene therapy. Technical considerations important to the exploration and pre-clinical development are presented, with an emphasis on vector technologies and delivery strategies whose capacity to generate and sustain functional transgene expression in vivo is well-established.
Collapse
Affiliation(s)
- Rachael S. Watson-Levings
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Glyn D. Palmer
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Padraic P. Levings
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - E. Anthony Dacanay
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Christopher H. Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MI, United States
| | - Steven C. Ghivizzani
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
- *Correspondence: Steven C. Ghivizzani,
| |
Collapse
|
26
|
Xu C, Sellke FW, Abid MR. Assessments of microvascular function in organ systems. Am J Physiol Heart Circ Physiol 2022; 322:H891-H905. [PMID: 35333121 PMCID: PMC9037705 DOI: 10.1152/ajpheart.00589.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 01/23/2023]
Abstract
Microvascular disease plays critical roles in the dysfunction of all organ systems, and there are many methods available to assess the microvasculature. These methods can either assess the target organ directly or assess an easily accessible organ such as the skin or retina so that inferences can be extrapolated to the other systems and/or related diseases. Despite the abundance of exploratory research on some of these modalities and their possible applications, there is a general lack of clinical use. This deficiency is likely due to two main reasons: the need for standardization of protocols to establish a role in clinical practice or the lack of therapies targeted toward microvascular dysfunction. Also, there remain some questions to be answered about the coronary microvasculature, as it is complex, heterogeneous, and difficult to visualize in vivo even with advanced imaging technology. This review will discuss novel approaches that are being used to assess microvasculature health in several key organ systems, and evaluate their clinical utility and scope for further development.
Collapse
Affiliation(s)
- Cynthia Xu
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Frank W Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - M Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
27
|
Oliveira Miranda C. Mesenchymal stem cells for lysosomal storage and polyglutamine disorders: Possible shared mechanisms. Eur J Clin Invest 2022; 52:e13707. [PMID: 34751953 DOI: 10.1111/eci.13707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/28/2021] [Accepted: 11/07/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Mesenchymal stem cells' (MSC) therapeutic potential has been investigated for the treatment of several neurodegenerative diseases. The fact these cells can mediate a beneficial effect in different neurodegenerative contexts strengthens their competence to target diverse mechanisms. On the other hand, distinct disorders may share similar mechanisms despite having singular neuropathological characteristics. METHODS We have previously shown that MSC can be beneficial for two disorders, one belonging to the groups of Lysosomal Storage Disorders (LSDs) - the Krabbe Disease or Globoid Cell Leukodystrophy, and the other to the family of Polyglutamine diseases (PolyQs) - the Machado-Joseph Disease or Spinocerebellar ataxia type 3. We gave also input into disease characterization since neuropathology and MSC's effects are intrinsically associated. This review aims at describing MSC's multimode of action in these disorders while emphasizing to possible mechanistic alterations they must share due to the accumulation of cellular toxic products. RESULTS Lysosomal storage disorders and PolyQs have different aetiology and associated symptoms, but both result from the accumulation of undegradable products inside neuronal cells due to inefficient clearance by the endosomal/lysosomal pathway. Moreover, numerous cellular mechanisms that become compromised latter are also shared by these two disease groups. CONCLUSIONS Here, we emphasize MSC's effect in improving proteostasis and autophagy cycling turnover, neuronal survival, synaptic activity and axonal transport. LSDs and PolyQs, though rare in their predominance, collectively affect many people and require our utmost dedication and efforts to get successful therapies due to their tremendous impact on patient s' lives and society.
Collapse
Affiliation(s)
- Catarina Oliveira Miranda
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Institute of Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
28
|
Bottomley MJ, Brook MO, Shankar S, Hester J, Issa F. Towards regulatory cellular therapies in solid organ transplantation. Trends Immunol 2022; 43:8-21. [PMID: 34844848 DOI: 10.1016/j.it.2021.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/10/2021] [Accepted: 11/01/2021] [Indexed: 01/03/2023]
Abstract
Organ transplantation is a modern medical success story. However, since its inception it has been limited by the need for pharmacological immunosuppression. Regulatory cellular therapies offer an attractive solution to these challenges by controlling transplant alloresponses through multiple parallel suppressive mechanisms. A number of cell types have seen an accelerated development into human trials and are now on the threshold of a long-awaited breakthrough in personalized transplant therapeutics. Here we assess recent developments with a focus on the most likely candidates, some of which have already facilitated successful immunosuppression withdrawal in early clinical trials. We propose that this may constitute a promising approach in clinical transplantation but also evaluate outstanding issues in the field, providing cause for cautious optimism.
Collapse
Affiliation(s)
- Matthew J Bottomley
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford Transplant Centre, Churchill Hospital, Oxford, UK
| | - Matthew O Brook
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford Transplant Centre, Churchill Hospital, Oxford, UK
| | - Sushma Shankar
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford Transplant Centre, Churchill Hospital, Oxford, UK
| | - Joanna Hester
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
29
|
Masterson CH, Ceccato A, Artigas A, Dos Santos C, Rocco PR, Rolandsson Enes S, Weiss DJ, McAuley D, Matthay MA, English K, Curley GF, Laffey JG. Mesenchymal stem/stromal cell-based therapies for severe viral pneumonia: therapeutic potential and challenges. Intensive Care Med Exp 2021; 9:61. [PMID: 34970706 PMCID: PMC8718182 DOI: 10.1186/s40635-021-00424-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/21/2021] [Indexed: 12/15/2022] Open
Abstract
Severe viral pneumonia is a significant cause of morbidity and mortality globally, whether due to outbreaks of endemic viruses, periodic viral epidemics, or the rarer but devastating global viral pandemics. While limited anti-viral therapies exist, there is a paucity of direct therapies to directly attenuate viral pneumonia-induced lung injury, and management therefore remains largely supportive. Mesenchymal stromal/stem cells (MSCs) are receiving considerable attention as a cytotherapeutic for viral pneumonia. Several properties of MSCs position them as a promising therapeutic strategy for viral pneumonia-induced lung injury as demonstrated in pre-clinical studies in relevant models. More recently, early phase clinical studies have demonstrated a reassuring safety profile of these cells. These investigations have taken on an added importance and urgency during the COVID-19 pandemic, with multiple trials in progress across the globe. In parallel with clinical translation, strategies are being investigated to enhance the therapeutic potential of these cells in vivo, with different MSC tissue sources, specific cellular products including cell-free options, and strategies to ‘licence’ or ‘pre-activate’ these cells, all being explored. This review will assess the therapeutic potential of MSC-based therapies for severe viral pneumonia. It will describe the aetiology and epidemiology of severe viral pneumonia, describe current therapeutic approaches, and examine the data suggesting therapeutic potential of MSCs for severe viral pneumonia in pre-clinical and clinical studies. The challenges and opportunities for MSC-based therapies will then be considered.
Collapse
Affiliation(s)
- C H Masterson
- Anaesthesia, School of Medicine, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - A Ceccato
- Intensive Care Unit, Hospital Universitari Sagrat Cor, Barcelona, Spain.,CIBER de Enfermedades Respiratorias (CIBERES), Sabbadell, Spain
| | - A Artigas
- CIBER de Enfermedades Respiratorias (CIBERES), Sabbadell, Spain.,Critical Center, Corporacion Sanitaria Universitaria Parc Tauli, Autonomous University of Barcelona, Sabadell, Spain
| | - C Dos Santos
- Keenan Center for Biomedical Research, St. Michael's Hospital, Bond St, Toronto, Canada.,Interdepartmental Division of Critical Care Medicine and Institutes of Medical Sciences, University of Toronto, Toronto, Canada
| | - P R Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - S Rolandsson Enes
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - D J Weiss
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - D McAuley
- Regional Intensive Care Unit, Royal Victoria Hospital, Belfast, UK.,Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - M A Matthay
- Department of Medicine and Anesthesia, University of California, San Francisco, CA, USA.,Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - K English
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - G F Curley
- Anaesthesia, School of Medicine, Royal College of Surgeons in Ireland, Dublin 9, Ireland
| | - J G Laffey
- Anaesthesia, School of Medicine, National University of Ireland, Galway, Ireland. .,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland. .,Department of Anaesthesia and Intensive Care Medicine, Galway University Hospitals, Saolta University Hospital Group, Galway, Ireland.
| |
Collapse
|
30
|
Bagno LL, Salerno AG, Balkan W, Hare JM. Mechanism of Action of Mesenchymal Stem Cells (MSCs): impact of delivery method. Expert Opin Biol Ther 2021; 22:449-463. [PMID: 34882517 DOI: 10.1080/14712598.2022.2016695] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Mesenchymal stromal cells (MSCs; AKA mesenchymal stem cells) stimulate healing and reduce inflammation. Promising therapeutic responses are seen in many late-phase clinical trials, but others have not satisfied their primary endpoints, making translation of MSCs into clinical practice difficult. These inconsistencies may be related to the route of MSC delivery, lack of product optimization, or varying background therapies received in clinical trials over time. AREAS COVERED Here we discuss the different routes of MSC delivery, highlighting the proposed mechanism(s) of therapeutic action as well as potential safety concerns. PubMed search criteria used: MSC plus: local administration; routes of administration; delivery methods; mechanism of action; therapy in different diseases. EXPERT OPINION Direct injection of MSCs using a controlled local delivery approach appears to have benefits in certain disease states, but further studies are required to make definitive conclusions regarding the superiority of one delivery method over another.
Collapse
Affiliation(s)
- Luiza L Bagno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alessandro G Salerno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami
| |
Collapse
|
31
|
Li A, Guo F, Pan Q, Chen S, Chen J, Liu HF, Pan Q. Mesenchymal Stem Cell Therapy: Hope for Patients With Systemic Lupus Erythematosus. Front Immunol 2021; 12:728190. [PMID: 34659214 PMCID: PMC8516390 DOI: 10.3389/fimmu.2021.728190] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/14/2021] [Indexed: 12/26/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease. Although previous studies have demonstrated that SLE is related to the imbalance of cells in the immune system, including B cells, T cells, and dendritic cells, etc., the mechanisms underlying SLE pathogenesis remain unclear. Therefore, effective and low side-effect therapies for SLE are lacking. Recently, mesenchymal stem cell (MSC) therapy for autoimmune diseases, particularly SLE, has gained increasing attention. This therapy can improve the signs and symptoms of refractory SLE by promoting the proliferation of Th2 and Treg cells and inhibiting the activity of Th1, Th17, and B cells, etc. However, MSC therapy is also reported ineffective in some patients with SLE, which may be related to MSC- or patient-derived factors. Therefore, the therapeutic effects of MSCs should be further confirmed. This review summarizes the status of MSC therapy in refractory SLE treatment and potential reasons for the ineffectiveness of MSC therapy from three perspectives. We propose various MSC modification methods that may be beneficial in enhancing the immunosuppression of MSCs in SLE. However, their safety and protective effects in patients with SLE still need to be confirmed by further experimental and clinical evidence.
Collapse
Affiliation(s)
- Aifen Li
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fengbiao Guo
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Quanren Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shuxian Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaxuan Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qingjun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
32
|
Masterson C, Gonzalez H, Laffey JG. Understanding the impact of the lung microenvironment to enhance the therapeutic potential of mesenchymal stromal cells for acute respiratory distress syndrome. Eur Respir J 2021; 58:58/4/2100986. [PMID: 34649970 DOI: 10.1183/13993003.00986-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 04/24/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Claire Masterson
- Anaesthesia, School of Medicine, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Hector Gonzalez
- Anaesthesia, School of Medicine, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - John G Laffey
- Anaesthesia, School of Medicine, National University of Ireland, Galway, Ireland .,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland.,Dept of Anaesthesia and Intensive Care Medicine, Galway University Hospitals, Saolta University Hospital Group, Galway, Ireland
| |
Collapse
|
33
|
Mesenchymal Stem Cells in the Treatment of COVID-19, a Promising Future. Cells 2021; 10:cells10102588. [PMID: 34685567 PMCID: PMC8533906 DOI: 10.3390/cells10102588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/11/2021] [Accepted: 09/17/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stem cells present in virtually all tissues; they have a potent self-renewal capacity and can differentiate into multiple cell types. They also affect the ambient tissue by the paracrine secretion of numerous factors in vivo, including the induction of other stem cells’ differentiation. In vitro, the culture media supernatant is named secretome and contains soluble molecules and extracellular vesicles that retain potent biological function in tissue regeneration. MSCs are considered safe for human treatment; their use does not involve ethical issues, as embryonic stem cells do not require genetic manipulation as induced pluripotent stem cells, and after intravenous injection, they are mainly found in the lugs. Therefore, these cells are currently being tested in various preclinical and clinical trials for several diseases, including COVID-19. Several affected COVID-19 patients develop induced acute respiratory distress syndrome (ARDS) associated with an uncontrolled inflammatory response. This condition causes extensive damage to the lungs and may leave serious post-COVID-19 sequelae. As the disease may cause systemic alterations, such as thromboembolism and compromised renal and cardiac function, the intravenous injection of MSCs may be a therapeutic alternative against multiple pathological manifestations. In this work, we reviewed the literature about MSCs biology, focusing on their function in pulmonary regeneration and their use in COVID-19 treatment.
Collapse
|