1
|
Da Silva K, Kumar P, Choonara YE. The paradigm of stem cell secretome in tissue repair and regeneration: Present and future perspectives. Wound Repair Regen 2025; 33:e13251. [PMID: 39780313 PMCID: PMC11711308 DOI: 10.1111/wrr.13251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
As the number of patients requiring organ transplants continues to rise exponentially, there is a dire need for therapeutics, with repair and regenerative properties, to assist in alleviating this medical crisis. Over the past decade, there has been a shift from conventional stem cell treatments towards the use of the secretome, the protein and factor secretions from cells. These components may possess novel druggable targets and hold the key to profoundly altering the field of regenerative medicine. Despite the progress in this field, clinical translation of secretome-containing products is limited by several challenges including but not limited to ensuring batch-to-batch consistency, the prevention of further heterogeneity, production of sufficient secretome quantities, product registration, good manufacturing practice protocols and the pharmacokinetic/pharmacodynamic profiles of all the components. Despite this, the secretome may hold the key to unlocking the regenerative blockage scientists have encountered for years. This review critically analyses the secretome derived from different cell sources and used in several tissues for tissue regeneration. Furthermore, it provides an overview of the current delivery strategies and the future perspectives for the secretome as a potential therapeutic. The success and possible shortcomings of the secretome are evaluated.
Collapse
Affiliation(s)
- Kate Da Silva
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
2
|
Vorstandlechner V, Copic D, Klas K, Direder M, Golabi B, Radtke C, Ankersmit HJ, Mildner M. The Secretome of Irradiated Peripheral Mononuclear Cells Attenuates Hypertrophic Skin Scarring. Pharmaceutics 2023; 15:pharmaceutics15041065. [PMID: 37111549 PMCID: PMC10143262 DOI: 10.3390/pharmaceutics15041065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Hypertrophic scars can cause pain, movement restrictions, and reduction in the quality of life. Despite numerous options to treat hypertrophic scarring, efficient therapies are still scarce, and cellular mechanisms are not well understood. Factors secreted by peripheral blood mononuclear cells (PBMCsec) have been previously described for their beneficial effects on tissue regeneration. In this study, we investigated the effects of PBMCsec on skin scarring in mouse models and human scar explant cultures at single-cell resolution (scRNAseq). Mouse wounds and scars, and human mature scars were treated with PBMCsec intradermally and topically. The topical and intradermal application of PBMCsec regulated the expression of various genes involved in pro-fibrotic processes and tissue remodeling. We identified elastin as a common linchpin of anti-fibrotic action in both mouse and human scars. In vitro, we found that PBMCsec prevents TGFβ-mediated myofibroblast differentiation and attenuates abundant elastin expression with non-canonical signaling inhibition. Furthermore, the TGFβ-induced breakdown of elastic fibers was strongly inhibited by the addition of PBMCsec. In conclusion, we conducted an extensive study with multiple experimental approaches and ample scRNAseq data demonstrating the anti-fibrotic effect of PBMCsec on cutaneous scars in mouse and human experimental settings. These findings point at PBMCsec as a novel therapeutic option to treat skin scarring.
Collapse
Affiliation(s)
- Vera Vorstandlechner
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Dragan Copic
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, 1090 Vienna, Austria
| | - Katharina Klas
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Martin Direder
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
- Department of Orthopedics and Trauma-Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Bahar Golabi
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Christine Radtke
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Hendrik J. Ankersmit
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence:
| |
Collapse
|
3
|
Copic D, Direder M, Schossleitner K, Laggner M, Klas K, Bormann D, Ankersmit HJ, Mildner M. Paracrine Factors of Stressed Peripheral Blood Mononuclear Cells Activate Proangiogenic and Anti-Proteolytic Processes in Whole Blood Cells and Protect the Endothelial Barrier. Pharmaceutics 2022; 14:pharmaceutics14081600. [PMID: 36015226 PMCID: PMC9415091 DOI: 10.3390/pharmaceutics14081600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 01/25/2023] Open
Abstract
Tissue-regenerative properties have been attributed to secreted paracrine factors derived from stem cells and other cell types. In particular, the secretome of γ-irradiated peripheral blood mononuclear cells (PBMCsec) has been shown to possess high tissue-regenerative and proangiogenic capacities in a variety of preclinical studies. In light of future therapeutic intravenous applications of PBMCsec, we investigated the possible effects of PBMCsec on white blood cells and endothelial cells lining the vasculature. To identify changes in the transcriptional profile, whole blood was drawn from healthy individuals and stimulated with PBMCsec for 8 h ex vivo before further processing for single-cell RNA sequencing. PBMCsec significantly altered the gene signature of granulocytes (17 genes), T-cells (45 genes), B-cells (72 genes), and, most prominently, monocytes (322 genes). We detected a strong upregulation of several tissue-regenerative and proangiogenic cyto- and chemokines in monocytes, including VEGFA, CXCL1, and CXCL5. Intriguingly, inhibitors of endopeptidase activity, such as SERPINB2, were also strongly induced. Measurement of the trans-endothelial electrical resistance of primary human microvascular endothelial cells revealed a strong barrier-protective effect of PBMCsec after barrier disruption. Together, we show that PBMCsec induces angiogenic and proteolytic processes in the blood and is able to attenuate endothelial barrier damage. These regenerative properties suggest that systemic application of PBMCsec might be a promising novel strategy to restore damaged organs.
Collapse
Affiliation(s)
- Dragan Copic
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria; (D.C.); (M.D.); (M.L.); (K.K.); (D.B.)
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Martin Direder
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria; (D.C.); (M.D.); (M.L.); (K.K.); (D.B.)
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Klaudia Schossleitner
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Maria Laggner
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria; (D.C.); (M.D.); (M.L.); (K.K.); (D.B.)
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Katharina Klas
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria; (D.C.); (M.D.); (M.L.); (K.K.); (D.B.)
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniel Bormann
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria; (D.C.); (M.D.); (M.L.); (K.K.); (D.B.)
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Hendrik Jan Ankersmit
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria; (D.C.); (M.D.); (M.L.); (K.K.); (D.B.)
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence: (H.J.A.); (M.M.)
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence: (H.J.A.); (M.M.)
| |
Collapse
|
4
|
Laggner M, Acosta GS, Kitzmüller C, Copic D, Gruber F, Altenburger LM, Vorstandlechner V, Gugerell A, Direder M, Klas K, Bormann D, Peterbauer A, Shibuya A, Bohle B, Ankersmit HJ, Mildner M. The secretome of irradiated peripheral blood mononuclear cells attenuates activation of mast cells and basophils. EBioMedicine 2022; 81:104093. [PMID: 35671621 PMCID: PMC9168057 DOI: 10.1016/j.ebiom.2022.104093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND IgE-mediated hypersensitivity is becoming increasingly prevalent and activation of mast cells and basophils represent key events in the pathophysiology of allergy. We have previously reported that the secretome of γ-irradiated peripheral blood mononuclear cells (PBMCsec) exerts beneficial anti-inflammatory effects. Yet, its ability to alleviate allergic symptoms has not been investigated so far. METHODS Several experimental in vitro and in vivo models have been used in this basic research study. A murine ear swelling model was used to study the effects of PBMCsec on 48/80-induced mast cell degranulation in vivo. The transcriptional profile of murine mast cells was analysed by single cell RNA sequencing (scRNAseq). Mast cell activation was studied in vitro using primary skin mast cells. Basophils from individuals allergic to birch pollens were used to investigate basophile activation by allergens. Transcriptomic and lipidomic analyses were used to identify mRNA expression and lipid species present in PBMCsec, respectively. FINDINGS Topical application of PBMCsec on mouse ears (C57BL/6) significantly reduced tissue swelling following intradermal injection of compound 48/80, an inducer of mast cell degranulation. Single cell RNA sequencing of PBMCsec-treated murine dermal mast cells (Balb/c) revealed a downregulation of genes involved in immune cell degranulation and Fc-receptor signalling. In addition, treatment of primary human dermal mast cells with PBMCsec strongly inhibited compound 48/80- and α-IgE-induced mediator release in vitro. Furthermore, PBMCsec remarkably attenuated allergen driven activation of basophils from allergic individuals. Transcriptomic analysis of these basophils showed that PBMCsec downregulated a distinct gene battery involved in immune cell degranulation and Fc-receptor signalling, corroborating results obtained from dermal mast cells. Finally, we identified the lipid fraction of PBMCsec as the major active ingredient involved in effector cell inhibition. INTERPRETATION Collectively, our data demonstrate that PBMCsec is able to reduce activation of mast cells and basophils, encouraging further studies on the potential use of PBMCsec for treating allergy. FUNDING Austrian Research Promotion Agency (852748 and 862068, 2015-2019), Vienna Business Agency (2343727, 2018-2020), Aposcience AG, Austrian Federal Ministry of Education, Science and Research (SPA06/055), Danube Allergy Research Cluster, Austrian Science Fund (I4437 and P32953).
Collapse
Affiliation(s)
- Maria Laggner
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria
| | - Gabriela Sánchez Acosta
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Claudia Kitzmüller
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Dragan Copic
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria
| | - Florian Gruber
- Department of Dermatology, Medical University of Vienna, Lazarettgasse 14, 1090 Vienna, Austria
| | | | - Vera Vorstandlechner
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria; Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Alfred Gugerell
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria
| | - Martin Direder
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria
| | - Katharina Klas
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria
| | - Daniel Bormann
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria
| | - Anja Peterbauer
- Aposcience AG, Vienna, Austria; Austrian Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria.
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Lazarettgasse 14, 1090 Vienna, Austria.
| |
Collapse
|
5
|
Secretome of Stressed Peripheral Blood Mononuclear Cells Alters Transcriptome Signature in Heart, Liver, and Spleen after an Experimental Acute Myocardial Infarction: An In Silico Analysis. BIOLOGY 2022; 11:biology11010116. [PMID: 35053121 PMCID: PMC8772778 DOI: 10.3390/biology11010116] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 12/21/2022]
Abstract
Simple Summary Acute myocardial infarction is characterized by impaired coronary blood flow, which leads to cardiac ischemia and, ultimately, compromised heart function. Damage and cellular responses are not limited to the non-perfused area, but rather affect the entire heart, as well as distal organs, such as the liver and spleen. We found that the therapeutic secretome of stressed white blood cells improved short-term and long-term cardiac performance in a porcine infarction model. In order to unravel the molecular events governing secretome-mediated tissue regeneration, we performed transcriptional analyses of the non-perfused, transition, and perfused heart, as well as the liver and spleen 24 h after myocardial infarction. We observed a highly tissue-specific effect of the secretome and, except for the transition zone, a uniform downregulation of pro-inflammatory factors and pathways. Simultaneously, the secretome strongly promoted the expression of genes that are essential for heart function in the non-perfused area. In the liver and spleen, different metabolic processes were induced. Together, our data suggest several plausible mechanisms by which the secretome improves heart function after cardiac ischemia. Deepening our understanding of the molecular processes identified here might uncover further pharmacologic strategies aiming at delimiting adverse cardiac remodeling and sequelae after myocardial infarction. Abstract Acute myocardial infarction (AMI) is a result of cardiac non-perfusion and leads to cardiomyocyte necrosis, inflammation, and compromised cardiac performance. Here, we showed that the secretome of γ-irradiated peripheral blood mononuclear cells (PBMCsec) improved heart function in a porcine AMI model and displayed beneficial long- and short-term effects. As an AMI is known to strongly affect gene regulation of the ischemia non-affected heart muscle and distal organs, we employed a transcriptomics approach to further study the immediate molecular events orchestrated using the PBMCsec in myocardium, liver, and spleen 24 h post ischemia. In the infarcted area, the PBMCsec mainly induced genes that were essential for cardiomyocyte function and simultaneously downregulated pro-inflammatory genes. Interestingly, genes associated with pro-inflammatory processes were activated in the transition zone, while being downregulated in the remote zone. In the liver, we observed a pronounced inhibition of immune responses using the PBMCsec, while genes involved in urea and tricarboxylic cycles were induced. The spleen displayed elevated lipid metabolism and reduced immunological processes. Together, our study suggested several types of pharmacodynamics by which the PBMCsec conferred immediate cardioprotection. Furthermore, our data supported the assumption that an AMI significantly affects distal organs, suggesting that a holistic treatment of an AMI, as achieved by PBMCsec, might be highly beneficial.
Collapse
|
6
|
Nozohouri S, Vaidya B, Abbruscato TJ. Exosomes in Ischemic Stroke. Curr Pharm Des 2021; 26:5533-5545. [PMID: 32534564 DOI: 10.2174/1381612826666200614180253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
Abstract
Ischemic stroke, a leading cause of mortality, results in severe neurological outcomes in the patients. Effective stroke therapies may significantly decrease the extent of injury. For this purpose, novel and efficient drug delivery strategies need to be developed. Among a myriad of therapeutic and drug delivery techniques, exosomes have shown promising results in ischemic stroke either by their intrinsic therapeutic characteristics, which can result in angiogenesis and neurogenesis or by acting as competent, biocompatible drug delivery vehicles to transport neurotherapeutic agents into the brain. In this review, we have discussed different methods of exosome isolation and cargo loading techniques, advantages and disadvantages of using exosomes as a drug delivery carrier and the therapeutic applications of exosomes with a focus on ischemic stroke therapy.
Collapse
Affiliation(s)
- Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| |
Collapse
|
7
|
Hsu TW, Lu YJ, Lin YJ, Huang YT, Hsieh LH, Wu BH, Lin YC, Chen LC, Wang HW, Chuang JC, Fang YQ, Huang CC. Transplantation of 3D MSC/HUVEC spheroids with neuroprotective and proangiogenic potentials ameliorates ischemic stroke brain injury. Biomaterials 2021; 272:120765. [PMID: 33780686 DOI: 10.1016/j.biomaterials.2021.120765] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 03/02/2021] [Accepted: 03/14/2021] [Indexed: 12/13/2022]
Abstract
Ischemic stroke, and the consequent brain cell death, is a common cause of death and disability worldwide. Current treatments that primarily aim to relieve symptoms are relatively inefficient in achieving brain tissue regeneration and functional recovery, and thus novel therapeutic options are urgently needed. Although cell-based therapies have shown promise for treating the infarcted brain, a recurring challenge is the inadequate retention and engraftment of transplanted cells at the target tissue, thereby limiting the ultimate therapeutic efficacy. Here, we show that transplantation of preassembled three-dimensional (3D) spheroids of mesenchymal stem cells (MSCs) and vascular endothelial cells (ECs) results in significantly improved cell retention and survival compared with conventional mixed-cell suspensions. The transplanted 3D spheroids exhibit notable neuroprotective, proneurogenic, proangiogenic and anti-scarring potential as evidenced by clear extracellular matrix structure formation and paracrine factor expression and secretion; this ultimately results in increased structural and motor function recovery in the brain of an ischemic stroke mouse model. Therefore, transplantation of MSCs and ECs using the 3D cell spheroid configuration not only reduces cell loss during cell harvesting/administration but also enhances the resultant therapeutic benefit, thus providing important proof-of-concept for future clinical translation.
Collapse
Affiliation(s)
- Ting-Wei Hsu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, 33305, Taiwan; Centre for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan; Centre for Biomedical Science and Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Jie Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Ting Huang
- College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Li-Hung Hsieh
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Bing-Huan Wu
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Li-Chi Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Hsin-Wen Wang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Jui-Che Chuang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yi-Qiao Fang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
8
|
Laggner M, Gugerell A, Copic D, Jeitler M, Springer M, Peterbauer A, Kremslehner C, Filzwieser-Narzt M, Gruber F, Madlener S, Erb M, Widder J, Lechner W, Georg D, Mildner M, Ankersmit HJ. Comparing the efficacy of γ- and electron-irradiation of PBMCs to promote secretion of paracrine, regenerative factors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:14-27. [PMID: 33768126 PMCID: PMC7960502 DOI: 10.1016/j.omtm.2021.02.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/19/2021] [Indexed: 11/28/2022]
Abstract
Cell-free secretomes represent a promising new therapeutic avenue in regenerative medicine, and γ-irradiation of human peripheral blood mononuclear cells (PBMCs) has been shown to promote the release of paracrine factors with high regenerative potential. Recently, the use of alternative irradiation sources, such as artificially generated β- or electron-irradiation, is encouraged by authorities. Since the effect of the less hazardous electron-radiation on the production and functions of paracrine factors has not been tested so far, we compared the effects of γ- and electron-irradiation on PBMCs and determined the efficacy of both radiation sources for producing regenerative secretomes. Exposure to 60 Gy γ-rays from a radioactive nuclide and 60 Gy electron-irradiation provided by a linear accelerator comparably induced cell death and DNA damage. The transcriptional landscapes of PBMCs exposed to either radiation source shared a high degree of similarity. Secretion patterns of proteins, lipids, and extracellular vesicles displayed similar profiles after γ- and electron-irradiation. Lastly, we detected comparable biological activities in functional assays reflecting the regenerative potential of the secretomes. Taken together, we were able to demonstrate that electron-irradiation is an effective, alternative radiation source for producing therapeutic, cell-free secretomes. Our study paves the way for future clinical trials employing secretomes generated with electron-irradiation in tissue-regenerative medicine.
Collapse
Affiliation(s)
- Maria Laggner
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria.,Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090 Vienna, Austria
| | - Alfred Gugerell
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria.,Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090 Vienna, Austria
| | - Dragan Copic
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria.,Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090 Vienna, Austria
| | - Markus Jeitler
- Core Facility Genomics, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Springer
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria.,Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090 Vienna, Austria
| | - Anja Peterbauer
- Austrian Red Cross Blood Transfusion Service of Upper Austria, 4020 Linz, Austria
| | - Christopher Kremslehner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria.,Christian Doppler Laboratory for Biotechnology of Skin Aging, 1090 Vienna, Austria
| | - Manuel Filzwieser-Narzt
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria.,Christian Doppler Laboratory for Biotechnology of Skin Aging, 1090 Vienna, Austria
| | - Florian Gruber
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria.,Christian Doppler Laboratory for Biotechnology of Skin Aging, 1090 Vienna, Austria
| | - Sibylle Madlener
- Molecular Neuro-Oncology, Department of Pediatrics and Adolescent Medicine, and Institute of Neurology, Medical University of Vienna, 1090 Vienna, Austria.,Comprehensive Cancer Center of the Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Erb
- SYNLAB Analytics and Services Switzerland AG, 4127 Birsfelden, Switzerland
| | - Joachim Widder
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Wolfgang Lechner
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Dietmar Georg
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Hendrik Jan Ankersmit
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria.,Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090 Vienna, Austria
| |
Collapse
|
9
|
Gugerell A, Gouya-Lechner G, Hofbauer H, Laggner M, Trautinger F, Almer G, Peterbauer-Scherb A, Seibold M, Hoetzenecker W, Dreschl C, Mildner M, Ankersmit HJ. Safety and clinical efficacy of the secretome of stressed peripheral blood mononuclear cells in patients with diabetic foot ulcer-study protocol of the randomized, placebo-controlled, double-blind, multicenter, international phase II clinical trial MARSYAS II. Trials 2021; 22:10. [PMID: 33407796 PMCID: PMC7789696 DOI: 10.1186/s13063-020-04948-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Background Diabetes and its sequelae such as diabetic foot ulcer are rising health hazards not only in western countries but all over the world. Effective, yet safe treatments are desperately sought for by physicians, healthcare providers, and of course patients. Methods/design APOSEC, a novel, innovative drug, is tested in the phase I/II study MARSYAS II, where its efficacy to promote healing of diabetic foot ulcers will be determined. To this end, the cell-free secretome of peripheral blood mononuclear cells (APOSEC) blended with a hydrogel will be applied topically three times weekly for 4 weeks. APOSEC is predominantly effective in hypoxia-induced tissue damages by modulating the immune system and enhancing angiogenesis, whereby its anti-microbial ability and neuro-regenerative capacity will exert further positive effects. In total, 132 patients will be enrolled in the multicenter, randomized, double-blind, placebo-controlled, parallel group, dose-ranging phase I/II study and treated with APOSEC at three dose levels or placebo for 4 weeks, followed by an 8-week follow-up period to evaluate safety and efficacy of the drug. Wound area reduction after 4 weeks of treatment will serve as the primary endpoint. Conclusion We consider our study protocol to be suitable to test topically administered APOSEC in patients suffering from diabetic foot ulcers in a clinical phase I/II trial. Trial registration EudraCT 2018-001653-27. Registered on 30 July 2019. ClinicalTrials.gov NCT04277598. Registered on 20 February 2020. Title: “A randomized, placebo-controlled, double-blind study to evaluate safety and dose-dependent clinical efficacy of APO-2 at three different doses in patients with diabetic foot ulcer (MARSYAS II)” Supplementary Information The online version contains supplementary material available at 10.1186/s13063-020-04948-1.
Collapse
Affiliation(s)
- Alfred Gugerell
- Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Aposcience AG, Vienna, Austria
| | | | - Helmut Hofbauer
- Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Aposcience AG, Vienna, Austria
| | - Maria Laggner
- Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Aposcience AG, Vienna, Austria
| | - Franz Trautinger
- Clinical Department for Skin and Venereal Diseases, Universitaetsklinikum St.Poelten, St. Poelten, Austria
| | | | | | - Marcus Seibold
- Aposcience AG, Vienna, Austria.,Austrian Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Wolfram Hoetzenecker
- Department of Dermatology and Venerology, Kepler University Hospital, Linz, Austria
| | - Christiane Dreschl
- Department of Surgery, Krankenhaus der Elisabethinen Klagenfurt, Klagenfurt, Austria
| | - Michael Mildner
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria. .,Aposcience AG, Vienna, Austria.
| |
Collapse
|
10
|
Hacker S, Mittermayr R, Traxler D, Keibl C, Resch A, Salminger S, Leiss H, Hacker P, Gabriel C, Golabi B, Pauzenberger R, Slezak P, Laggner M, Mildner M, Michlits W, Ankersmit HJ. The secretome of stressed peripheral blood mononuclear cells increases tissue survival in a rodent epigastric flap model. Bioeng Transl Med 2021; 6:e10186. [PMID: 33532586 PMCID: PMC7823127 DOI: 10.1002/btm2.10186] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022] Open
Abstract
Reconstructive surgery transfers viable tissue to cover defects and to restore aesthetic and functional properties. Failure rates after free flap surgery range from 3 to 7%. Co-morbidities such as diabetes mellitus or peripheral vascular disease increase the risk of flap failure up to 4.5-fold. Experimental therapeutic concepts commonly use a monocausal approach by applying single growth factors. The secretome of γ-irradiated, stressed peripheral blood mononuclear cells (PBMCsec) resembles the physiological environment necessary for tissue regeneration. Its application led to improved wound healing rates and a two-fold increase in blood vessel counts in previous animal models. We hypothesized that PBMCsec has beneficial effects on the survival of compromised flap tissue by reducing the necrosis rate and increasing angiogenesis. Surgery was performed on 39 male Sprague-Dawley rats (control, N = 13; fibrin sealant, N = 14; PBMCsec, N = 12). PBMCsec was produced according to good manufacturing practices (GMP) guidelines and 2 ml were administered intraoperatively at a concentration of 2.5 × 107 cells/ml using fibrin sealant as carrier substance. Flap perfusion and necrosis (as percentage of the total flap area) were analyzed using Laser Doppler Imaging and digital image planimetry on postoperative days 3 and 7. Immunohistochemical stainings for von Willebrand factor (vWF) and Vascular Endothelial Growth Factor-receptor-3 (Flt-4) were performed on postoperative day 7 to evaluate formation of blood vessels and lymphatic vessels. Seroma formation was quantified using a syringe and flap adhesion and tissue edema were evaluated clinically through a cranial incision by a blinded observer according to previously described criteria on postoperative day 7. We found a significantly reduced tissue necrosis rate (control: 27.8% ± 8.6; fibrin: 22.0% ± 6.2; 20.9% reduction, p = .053 vs. control; PBMCsec: 19.1% ± 7.2; 31.1% reduction, p = .012 vs. control; 12.9% reduction, 0.293 vs. fibrin) together with increased vWF+ vessel counts (control: 70.3 ± 16.3 vessels/4 fields at 200× magnification; fibrin: 67.8 ± 12.1; 3.6% reduction, p = .651, vs. control; PBMCsec: 85.9 ± 20.4; 22.2% increase, p = .045 vs. control; 26.7% increase, p = .010 vs. fibrin) on postoperative day 7 after treatment with PBMCsec. Seroma formation was decreased after treatment with fibrin sealant with or without the addition of PBMCsec. (control: 11.9 ± 9.7 ml; fibrin: 1.7 ± 5.3, 86.0% reduction, 0.004 vs. control; PBMCsec: 0.6 ± 2.0; 94.8% reduction, p = .001 vs. control; 62.8% reduction, p = .523 vs. fibrin). We describe the beneficial effects of a secretome derived from γ-irradiated PBMCs on tissue survival, angiogenesis, and clinical parameters after flap surgery in a rodent epigastric flap model.
Collapse
Affiliation(s)
- Stefan Hacker
- Division of Plastic and Reconstructive SurgeryMedical University of ViennaViennaAustria
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and RegenerationViennaAustria
| | - Rainer Mittermayr
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyViennaAustria
| | - Denise Traxler
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and RegenerationViennaAustria
| | - Claudia Keibl
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyViennaAustria
| | - Annika Resch
- Division of Plastic and Reconstructive SurgeryMedical University of ViennaViennaAustria
| | - Stefan Salminger
- Division of Plastic and Reconstructive SurgeryMedical University of ViennaViennaAustria
| | - Harald Leiss
- Division of RheumatologyMedical University of ViennaViennaAustria
| | - Philipp Hacker
- Department of Oral‐ and Maxillofacial SurgeryUniversity Clinic Sankt PoeltenSankt PoeltenAustria
| | - Christian Gabriel
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyViennaAustria
- Department of Blood Group Serology and Transfusion MedicineMedical University of GrazAustria
| | - Bahar Golabi
- Department of DermatologyMedical University of ViennaViennaAustria
| | - Reinhard Pauzenberger
- Division of Plastic and Reconstructive SurgeryMedical University of ViennaViennaAustria
| | - Paul Slezak
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyViennaAustria
| | - Maria Laggner
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and RegenerationViennaAustria
| | - Michael Mildner
- Department of DermatologyMedical University of ViennaViennaAustria
| | - Wolfgang Michlits
- Department of Plastic and Reconstructive SurgeryHospital Wiener NeustadtWiener NeustadtAustria
| | - Hendrik J. Ankersmit
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and RegenerationViennaAustria
- Division of Thoracic SurgeryMedical University of ViennaViennaAustria
| |
Collapse
|
11
|
TGF-β in the Secretome of Irradiated Peripheral Blood Mononuclear Cells Supports In Vitro Osteoclastogenesis. Int J Mol Sci 2020; 21:ijms21228569. [PMID: 33202935 PMCID: PMC7696998 DOI: 10.3390/ijms21228569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Osteoclastogenesis required for bone remodeling is also a key pathologic mechanism of inflammatory osteolysis being controlled by paracrine factors released from dying cells. The secretome of irradiated, dying peripheral blood mononuclear cells (PBMCs) has a major impact on the differentiation of myeloid cells into dendritic cells, and macrophage polarization. The impact on osteoclastogenesis, however, has not been reported. For this aim, we used murine bone marrow macrophages exposed to RANKL and M-CSF to initiate osteoclastogenesis, with and without the secretome obtained from γ-irradiated PBMCs. We reported that the secretome significantly enhanced in vitro osteoclastogenesis as determined by means of histochemical staining of the tartrate-resistant acid phosphatase (TRAP), as well as the expression of the respective target genes, including TRAP and cathepsin K. Considering that TGF-β enhanced osteoclastogenesis, we confirmed the TGF-β activity in the secretome with a bioassay that was based on the increased expression of IL11 in fibroblasts. Neutralizing TGF-β by an antibody decreased the ability of the secretome to support osteoclastogenesis. These findings suggested that TGF-β released by irradiated PBMCs could enhance the process of osteoclastogenesis in vitro.
Collapse
|
12
|
Haque N, Fareez IM, Fong LF, Mandal C, Kasim NHA, Kacharaju KR, Soesilawati P. Role of the CXCR4-SDF1-HMGB1 pathway in the directional migration of cells and regeneration of affected organs. World J Stem Cells 2020. [DOI: 10.4252/wjsc.v12.i9.0000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
13
|
Haque N, Fareez IM, Fong LF, Mandal C, Abu Kasim NH, Kacharaju KR, Soesilawati P. Role of the CXCR4-SDF1-HMGB1 pathway in the directional migration of cells and regeneration of affected organs. World J Stem Cells 2020; 12:938-951. [PMID: 33033556 PMCID: PMC7524697 DOI: 10.4252/wjsc.v12.i9.938] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/18/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
In recent years, several studies have reported positive outcomes of cell-based therapies despite insufficient engraftment of transplanted cells. These findings have created a huge interest in the regenerative potential of paracrine factors released from transplanted stem or progenitor cells. Interestingly, this notion has also led scientists to question the role of proteins in the secretome produced by cells, tissues or organisms under certain conditions or at a particular time of regenerative therapy. Further studies have revealed that the secretomes derived from different cell types contain paracrine factors that could help to prevent apoptosis and induce proliferation of cells residing within the tissues of affected organs. This could also facilitate the migration of immune, progenitor and stem cells within the body to the site of inflammation. Of these different paracrine factors present within the secretome, researchers have given proper consideration to stromal cell-derived factor-1 (SDF1) that plays a vital role in tissue-specific migration of the cells needed for regeneration. Recently researchers recognized that SDF1 could facilitate site-specific migration of cells by regulating SDF1-CXCR4 and/or HMGB1-SDF1-CXCR4 pathways which is vital for tissue regeneration. Hence in this study, we have attempted to describe the role of different types of cells within the body in facilitating regeneration while emphasizing the HMGB1-SDF1-CXCR4 pathway that orchestrates the migration of cells to the site where regeneration is needed.
Collapse
Affiliation(s)
- Nazmul Haque
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Selangor 42610, Malaysia
| | - Ismail M Fareez
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Selangor 42610, Malaysia
| | - Liew Fong Fong
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Selangor 42610, Malaysia
| | - Chanchal Mandal
- Biotechnology and Genetic Engineering Discipline, Life Science, Khulna University, Khulna 9208, Bangladesh
| | - Noor Hayaty Abu Kasim
- Faculty of Dentistry, University Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
- Faculty of Dental Medicine, Universitas Airlangga, Surabaya 411007, Indonesia
| | - Kranthi Raja Kacharaju
- Department of Conservative Dentistry, Faculty of Dentistry MAHSA University, Selangor 42610, Malaysia
| | - Pratiwi Soesilawati
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya 60115, Indonesia
| |
Collapse
|
14
|
Mangin G, Kubis N. Cell Therapy for Ischemic Stroke: How to Turn a Promising Preclinical Research into a Successful Clinical Story. Stem Cell Rev Rep 2020; 15:176-193. [PMID: 30443706 DOI: 10.1007/s12015-018-9864-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Stroke is a major public health issue with limited treatment. The pharmacologically or mechanically removing of the clot is accessible to less than 10% of the patients. Stem cell therapy is a promising alternative strategy since it increases the therapeutic time window but many issues remain unsolved. To avoid a new dramatic failure when translating experimental data on the bedside, this review aims to highlight the indispensable checkpoints to make a successful clinical trial based on the current preclinical literature. The large panel of progenitors/ stem cells at the researcher's disposal is to be used wisely, regarding the type of cells, the source of cells, the route of delivery, the time window, since it will directly affect the outcome. Mechanisms are still incompletely understood, although recent studies have focused on the inflammation modulation of most cells types.
Collapse
Affiliation(s)
| | - Nathalie Kubis
- INSERM U965, F-75475, Paris, France. .,Sorbonne Paris Cité, Université Paris Diderot, F-75475, Paris, France. .,Service de Physiologie Clinique-Explorations Fonctionnelles, AP-HP, Hôpital Lariboisière, 2 rue Ambroise Paré, F-75475, Paris, France.
| |
Collapse
|
15
|
Panahipour L, Kochergina E, Laggner M, Zimmermann M, Mildner M, Ankersmit HJ, Gruber R. Role for Lipids Secreted by Irradiated Peripheral Blood Mononuclear Cells in Inflammatory Resolution in Vitro. Int J Mol Sci 2020; 21:ijms21134694. [PMID: 32630157 PMCID: PMC7370068 DOI: 10.3390/ijms21134694] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 06/24/2020] [Accepted: 06/28/2020] [Indexed: 12/18/2022] Open
Abstract
Periodontal inflammation is associated with dying cells that potentially release metabolites helping to promote inflammatory resolution. We had shown earlier that the secretome of irradiated, dying peripheral blood mononuclear cells support in vitro angiogenesis. However, the ability of the secretome to promote inflammatory resolution remains unknown. Here, we determined the expression changes of inflammatory cytokines in murine bone marrow macrophages, RAW264.7 cells, and gingival fibroblasts exposed to the secretome obtained from γ-irradiated peripheral blood mononuclear cells in vitro by RT-PCR and immunoassays. Nuclear translocation of p65 was detected by immunofluorescence staining. Phosphorylation of p65 and degradation of IκB was determined by Western blot. The secretome of irradiated peripheral blood mononuclear cells significantly decreased the expression of IL1 and IL6 in primary macrophages and RAW264.7 cells when exposed to LPS or saliva, and of IL1, IL6, and IL8 in gingival fibroblasts when exposed to IL-1β and TNFα. These changes were associated with decreased phosphorylation and nuclear translocation of p65 but not degradation of IκB in macrophages. We also show that the lipid fraction of the secretome lowered the inflammatory response of macrophages exposed to the inflammatory cues. These results demonstrate that the secretome of irradiated peripheral blood mononuclear cells can lower an in vitro simulated inflammatory response, supporting the overall concept that the secretome of dying cells promotes inflammatory resolution.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (E.K.)
| | - Evgeniya Kochergina
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (E.K.)
| | - Maria Laggner
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Währingergürtel 18-20, 1090 Vienna, Austria; (M.L.); (H.J.A.)
- Division of Thoracic Surgery, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria
| | - Matthias Zimmermann
- Department of Oral and Maxillofacial Surgery, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria;
| | - Michael Mildner
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria;
| | - Hendrik J. Ankersmit
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Währingergürtel 18-20, 1090 Vienna, Austria; (M.L.); (H.J.A.)
- Division of Thoracic Surgery, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (E.K.)
- Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1200 Vienna, Austria
- Correspondence:
| |
Collapse
|
16
|
Laggner M, Copic D, Nemec L, Vorstandlechner V, Gugerell A, Gruber F, Peterbauer A, Ankersmit HJ, Mildner M. Therapeutic potential of lipids obtained from γ-irradiated PBMCs in dendritic cell-mediated skin inflammation. EBioMedicine 2020; 55:102774. [PMID: 32403085 PMCID: PMC7218268 DOI: 10.1016/j.ebiom.2020.102774] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Since numerous pathological conditions are evoked by unwanted dendritic cell (DC) activity, therapeutic agents modulating DC functions are of great medical interest. In regenerative medicine, cellular secretomes have gained increasing attention and valuable immunomodulatory properties have been attributed to the secretome of γ-irradiated peripheral blood mononuclear cells (PBMCs). Potential effects of the PBMC secretome (PBMCsec) on key DC functions have not been elucidated so far. METHODS We used a hapten-mediated murine model of contact hypersensitivity (CH) to study the effects of PBMCsec on DCs in vivo. Effects of PBMCsec on human DCs were investigated in monocyte-derived DCs (MoDC) and ex vivo skin cultures. DCs were phenotypically characterised by transcriptomics analyses and flow cytometry. DC function was evaluated by cytokine secretion, antigen uptake, PBMC proliferation and T-cell priming. FINDINGS PBMCsec significantly alleviated tissue inflammation and cellular infiltration in hapten-sensitized mice. We found that PBMCsec abrogated differentiation of MoDCs, indicated by lower expression of classical DC markers CD1a, CD11c and MHC class II molecules. Furthermore, PBMCsec reduced DC maturation, antigen uptake, lipopolysaccharides-induced cytokine secretion, and DC-mediated immune cell proliferation. Moreover, MoDCs differentiated with PBMCsec displayed diminished ability to prime naïve CD4+T-cells into TH1 and TH2 cells. Furthermore, PBMCsec modulated the phenotype of DCs present in the skin in situ. Mechanistically, we identified lipids as the main biomolecule accountable for the observed immunomodulatory effects. INTERPRETATION Together, our data describe DC-modulatory actions of lipids secreted by stressed PBMCs and suggest PBMCsec as a therapeutic option for treatment of DC-mediated inflammatory skin conditions. FUNDING This research project was supported by the Austrian Research Promotion Agency (Vienna, Austria; grant "APOSEC" 862068; 2015-2019) and the Vienna Business Agency (Vienna, Austria; grant "APOSEC to clinic" 2343727).
Collapse
Affiliation(s)
- Maria Laggner
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Vienna, Austria; Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Dragan Copic
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Vienna, Austria; Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Lucas Nemec
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Lazarettgasse 14, 1090 Vienna, Austria
| | - Vera Vorstandlechner
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Vienna, Austria; Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Alfred Gugerell
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Vienna, Austria; Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Florian Gruber
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Lazarettgasse 14, 1090 Vienna, Austria
| | - Anja Peterbauer
- Austrian Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Hendrik J Ankersmit
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Vienna, Austria; Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| | - Michael Mildner
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Lazarettgasse 14, 1090 Vienna, Austria.
| |
Collapse
|
17
|
Laggner M, Gugerell A, Bachmann C, Hofbauer H, Vorstandlechner V, Seibold M, Gouya Lechner G, Peterbauer A, Madlener S, Demyanets S, Sorgenfrey D, Ostler T, Erb M, Mildner M, Ankersmit HJ. Reproducibility of GMP-compliant production of therapeutic stressed peripheral blood mononuclear cell-derived secretomes, a novel class of biological medicinal products. Stem Cell Res Ther 2020; 11:9. [PMID: 31900195 PMCID: PMC6942406 DOI: 10.1186/s13287-019-1524-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/29/2019] [Accepted: 12/10/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The recent concept of secretome-based tissue regeneration has profoundly altered the field of regenerative medicine and offers promising novel therapeutic options. In contrast to medicinal products with a single active substance, cell-derived secretomes comprise pleiotropic bioactive ingredients, representing a major obstacle for reproducible drug product efficacy and warranting patient safety. Good manufacturing practice (GMP)-compliant production guarantees high batch-to-batch consistency and reproducible efficacy of biological medicinal products, but different batches of cellular secretomes produced under GMP have not been compared yet, and suitable quality control parameters have not been established. To this end, we analyzed diverse biological and functional parameters of different batches produced under GMP of the secretome obtained from γ-irradiated peripheral blood mononuclear cells with proven tissue regenerative properties in infarcted myocardium, stroke, spinal cord injury, and skin wounds. METHODS We quantified key secretome ingredients, including cytokines, lipids, and extracellular vesicles, and functionally assessed potency in tube formation assay, ex vivo aortic ring sprouting assay, and cell-based protein and reporter gene assays. Furthermore, we determined secretome stability in different batches after 6 months of storage at various ambient temperatures. RESULTS We observed that inter-batch differences in the bioactive components and secretome properties were small despite considerable differences in protein concentrations and potencies between individual donor secretomes. Stability tests showed that the analytical and functional properties of the secretomes remained stable when lyophilisates were stored at temperatures up to + 5 °C for 6 months. CONCLUSIONS We are the first to demonstrate the consistent production of cell-derived, yet cell-free secretome as a biological medicinal product. The results from this study provide the basis for selecting appropriate quality control parameters for GMP-compliant production of therapeutic cell secretomes and pave the way for future clinical trials employing secretomes in tissue regenerative medicine.
Collapse
Affiliation(s)
- Maria Laggner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Aposcience AG, Vienna, Austria
| | - Alfred Gugerell
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Aposcience AG, Vienna, Austria
| | | | - Helmut Hofbauer
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Aposcience AG, Vienna, Austria
| | - Vera Vorstandlechner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Aposcience AG, Vienna, Austria
| | | | | | - Anja Peterbauer
- Austrian Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Sibylle Madlener
- Molecular Neuro-Oncology, Department of Pediatrics and Adolescent Medicine and Institute of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria
| | - Svitlana Demyanets
- Department for Laboratory Medicine at the Medical University of Vienna, Vienna, Austria
| | | | - Tobias Ostler
- SYNLAB Analytics and Services Switzerland AG, Birsfelden, Switzerland
| | - Michael Erb
- SYNLAB Analytics and Services Switzerland AG, Birsfelden, Switzerland
| | - Michael Mildner
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.
- Aposcience AG, Vienna, Austria.
| |
Collapse
|
18
|
Gugerell A, Sorgenfrey D, Laggner M, Raimann J, Peterbauer A, Bormann D, Suessner S, Gabriel C, Moser B, Ostler T, Mildner M, Ankersmit HJ. Viral safety of APOSECTM: a novel peripheral blood mononuclear cell derived-biological for regenerative medicine. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2020; 18:30-39. [PMID: 30865581 PMCID: PMC7053523 DOI: 10.2450/2019.0249-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 01/31/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Viral reduction and inactivation of cell-derived biologicals is paramount for patients' safety and so viral reduction needs to be demonstrated to regulatory bodies in order to obtain marketing authorisation. Allogeneic human blood-derived biological medicinal products require special attention. APOSECTM, the secretome harvested from selected human blood cells, is a new biological with promising regenerative capabilities. We evaluated the effectiveness of inactivation of model viruses by methylene blue/light treatment, lyophilisation, and gamma irradiation during the manufacturing process of APOSECTM. MATERIALS AND METHODS Samples of intermediates of APOSECTM were acquired during the manufacturing process and spiked with bovine viral diarrhoea virus (BVDV), human immunodeficiency virus type 1 (HIV-1), pseudorabies virus (PRV), hepatitis A virus (HAV), and porcine parvovirus (PPV). Viral titres were assessed with suitable cell lines. RESULTS Methylene blue-assisted viral reduction is mainly effective against enveloped viruses: the minimum log10 reduction factors for BVDV, HIV-1, and PRV were ≥6.42, ≥6.88, and ≥6.18, respectively, with no observed residual infectivity. Viral titres of both HAV and PPV were not significantly reduced, indicating minor inactivation of non-enveloped viruses. Lyophilisation had minor effects on the viability of several enveloped model viruses. Gamma irradiation contributes to the viral safety by reduction of enveloped viruses (BVDV: ≥2.42; HIV-1: 4.53; PRV: ≥4.61) and to some degree of non-enveloped viruses as seen for HAV with a minimum log10 reduction factor of 2.92. No significant reduction could be measured for the non-enveloped virus PPV (2.60). DISCUSSION Three manufacturing steps of APOSECTM were evaluated under Good Laboratory Practice conditions for their efficacy at reducing and inactivating potentially present viruses. It could be demonstrated that all three steps contribute to the viral safety of APOSECTM.
Collapse
Affiliation(s)
- Alfred Gugerell
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | | | - Maria Laggner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Jürgen Raimann
- Charles River Laboratories Germany GmbH (CRL), Cologne, Germany
| | - Anja Peterbauer
- Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Daniel Bormann
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Susanne Suessner
- Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Christian Gabriel
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Bernhard Moser
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Tobias Ostler
- SYNLAB Analytics and Services Switzerland AG, Birsfelden, Switzerland
| | - Michael Mildner
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Hendrik J Ankersmit
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Wiener Wirtschaftsagentur Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
- Austrian Research Promotion Agency FFG Projects 852748 and 862068 "APOSEC", Medical University Vienna, Vienna, Austria
| |
Collapse
|
19
|
Bonova P, Jachova J, Nemethova M, Macakova L, Bona M, Gottlieb M. Rapid remote conditioning mediates modulation of blood cell paracrine activity and leads to the production of a secretome with neuroprotective features. J Neurochem 2019; 154:99-111. [DOI: 10.1111/jnc.14889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/03/2019] [Accepted: 10/07/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Petra Bonova
- Institute of Neurobiology Biomedical Research Center of Slovak Academy of Sciences Kosice Slovak Republic
| | - Jana Jachova
- Institute of Neurobiology Biomedical Research Center of Slovak Academy of Sciences Kosice Slovak Republic
| | - Miroslava Nemethova
- Institute of Neurobiology Biomedical Research Center of Slovak Academy of Sciences Kosice Slovak Republic
| | - Lubica Macakova
- Institute of Neurobiology Biomedical Research Center of Slovak Academy of Sciences Kosice Slovak Republic
| | - Martin Bona
- Department of Medical Physiology Faculty of Medicine Pavol Jozef Safarik University in Kosice Kosice Slovak Republic
| | - Miroslav Gottlieb
- Institute of Neurobiology Biomedical Research Center of Slovak Academy of Sciences Kosice Slovak Republic
| |
Collapse
|
20
|
Simader E, Beer L, Laggner M, Vorstandlechner V, Gugerell A, Erb M, Kalinina P, Copic D, Moser D, Spittler A, Tschachler E, Jan Ankersmit H, Mildner M. Tissue-regenerative potential of the secretome of γ-irradiated peripheral blood mononuclear cells is mediated via TNFRSF1B-induced necroptosis. Cell Death Dis 2019; 10:729. [PMID: 31570701 PMCID: PMC6768878 DOI: 10.1038/s41419-019-1974-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/05/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022]
Abstract
Peripheral blood mononuclear cells (PBMCs) have been shown to produce and release a plethora of pro-angiogenetic factors in response to γ-irradiation, partially accounting for their tissue-regenerative capacity. Here, we investigated whether a certain cell subtype of PBMCs is responsible for this effect, and whether the type of cell death affects the pro-angiogenic potential of bioactive molecules released by γ-irradiated PBMCs. PBMCs and PBMC subpopulations, including CD4+ and CD8+ T cells, B cells, monocytes, and natural killer cells, were isolated and subjected to high-dose γ-irradiation. Transcriptome analysis revealed subpopulation-specific responses to γ-irradiation with distinct activation of pro-angiogenic pathways, cytokine production, and death receptor signalling. Analysis of the proteins released showed that interactions of the subsets are important for the generation of a pro-angiogenic secretome. This result was confirmed at the functional level by the finding that the secretome of γ-irradiated PBMCs displayed higher pro-angiogenic activity in an aortic ring assay. Scanning electron microscopy and image stream analysis of γ-irradiated PBMCs revealed distinct morphological changes, indicative for apoptotic and necroptotic cell death. While inhibition of apoptosis had no effect on the pro-angiogenic activity of the secretome, inhibiting necroptosis in stressed PBMCs abolished blood vessel sprouting. Mechanistically, we identified tumor necrosis factor (TNF) receptor superfamily member 1B as the main driver of necroptosis in response to γ-irradiation in PBMCs, which was most likely mediated via membrane-bound TNF-α. In conclusion, our study demonstrates that the pro-angiogenic activity of the secretome of γ-irradiated PBMCs requires interplay of different PBMC subpopulations. Furthermore, we show that TNF-dependent necroptosis is an indispensable molecular process for conferring tissue-regenerative activity and for the pro-angiogenic potential of the PBMC secretome. These findings contribute to a better understanding of secretome-based therapies in regenerative medicine.
Collapse
Affiliation(s)
- Elisabeth Simader
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.,Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria
| | - Lucian Beer
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.,Department of Radiology and Cancer Research UK Cambridge Center, Cambridge, CB2 0QQ, UK
| | - Maria Laggner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Vera Vorstandlechner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Alfred Gugerell
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Michael Erb
- Synlab Analytics and Services Switzerland AG, Birsfelden, Switzerland
| | - Polina Kalinina
- Research Division of Biology and Pathobiology of the SkinDepartment of Dermatology, Research Division of Biology and Pathobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Dragan Copic
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Doris Moser
- Division of Oral and Maxillofacial Surgery, Medical University of Vienna, Vienna, Austria
| | - Andreas Spittler
- Research Laboratories, Core Facility Flow Cytometry, Medical University of Vienna, Vienna, Austria
| | - Erwin Tschachler
- Research Division of Biology and Pathobiology of the SkinDepartment of Dermatology, Research Division of Biology and Pathobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria. .,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria. .,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria.
| | - Michael Mildner
- Research Division of Biology and Pathobiology of the SkinDepartment of Dermatology, Research Division of Biology and Pathobiology of the Skin, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
21
|
Dabrowska S, Andrzejewska A, Lukomska B, Janowski M. Neuroinflammation as a target for treatment of stroke using mesenchymal stem cells and extracellular vesicles. J Neuroinflammation 2019; 16:178. [PMID: 31514749 PMCID: PMC6743114 DOI: 10.1186/s12974-019-1571-8] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke is the third cause of death in the developed countries and the main reason of severe disability. Brain ischemia leads to the production of damage-associated molecular patterns (DAMPs) by neurons and glial cells which results in astrocyte and microglia activation, pro-inflammatory cytokines and chemokines production, blood-brain barrier (BBB) disruption, infiltration of leukocytes from the peripheral blood into the infarcted area, and further exacerbation of tissue damage. However, some immune cells such as microglia or monocytes are capable to change their phenotype to anti-inflammatory, produce anti-inflammatory cytokines, and protect injured nervous tissue. In this situation, therapies, which will modulate the immune response after brain ischemia, such as transplantation of mesenchymal stem cells (MSCs) are catching interest. Many experimental studies of ischemic stroke revealed that MSCs are able to modulate immune response and act neuroprotective, through stimulation of neurogenesis, oligodendrogenesis, astrogenesis, and angiogenesis. MSCs may also have an ability to replace injured cells, but the release of paracrine factors directly into the environment or via extracellular vesicles (EVs) seems to play the most pronounced role. EVs are membrane structures containing proteins, lipids, and nucleic acids, and they express similar properties as the cells from which they are derived. However, EVs have lower immunogenicity, do not express the risk of vessel blockage, and have the capacity to cross the blood-brain barrier. Experimental studies of ischemic stroke showed that EVs have immunomodulatory and neuroprotective properties; therefore, they can stimulate neurogenesis and angiogenesis. Up to now, 20 clinical trials with MSC transplantation into patients after stroke were performed, from which two concerned on only hemorrhagic stroke and 13 studied only on ischemic stroke. There is no clinical trial with EV injection into patients after brain ischemia so far, but the case with miR-124-enriched EVs administration is planned and probably there will be more clinical studies with EV transplantation in the near future.
Collapse
Affiliation(s)
- Sylwia Dabrowska
- NeuroRepair Department, Mossakowski Medical Research Centre, PAS, 5 Pawinskiego Street, 02-106, Warsaw, Poland
| | - Anna Andrzejewska
- NeuroRepair Department, Mossakowski Medical Research Centre, PAS, 5 Pawinskiego Street, 02-106, Warsaw, Poland
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Centre, PAS, 5 Pawinskiego Street, 02-106, Warsaw, Poland
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, HSF III, 620 W. Baltimore street, Baltimore, MD, 21201, USA.
| |
Collapse
|
22
|
Zhu Z, Zheng L, Li Y, Huang T, Chao YC, Pan L, Zhu H, Zhao Y, Yu W, Li P. Potential Immunotherapeutic Targets on Myeloid Cells for Neurovascular Repair After Ischemic Stroke. Front Neurosci 2019; 13:758. [PMID: 31447626 PMCID: PMC6696904 DOI: 10.3389/fnins.2019.00758] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/08/2019] [Indexed: 12/11/2022] Open
Abstract
Neurological deficits and cognitive dysfunctions caused by acute ischemic stroke pose enormous burden to the stroke families and the communities. Restoration of the normal function of the neurovascular unit following ischemic stroke is critical for improving neurological recovery and cognitive functions after stroke. Recent evidence suggests that the myeloid cells including both the resident microglia and infiltrating monocytes/macrophages and neutrophils are highly plastic in response to the environmental cues. They intimately interact with multiple components of the neurovascular unit in response to the alarmins, danger associated pattern molecules (DAMPs) and other signals released from the ischemic brain. The aim of this review is to discuss the reciprocal interactions between the myeloid cells and the ischemic neurovascular unit during the late repair phase of cerebral ischemic stroke. We also summarize potential immunotherapeutic targets on myeloid cells and new therapeutic approaches targeting myeloid cells, such as cell transplantation, mitochondrial dynamic and extracellular vesicles-based therapy et al to enhance neurovascular repair for better stroke recovery.
Collapse
Affiliation(s)
- Ziyu Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Li Zheng
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tingting Huang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yu-Chieh Chao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lijun Pan
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hui Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yanhua Zhao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
23
|
Wuschko S, Gugerell A, Chabicovsky M, Hofbauer H, Laggner M, Erb M, Ostler T, Peterbauer A, Suessner S, Demyanets S, Leuschner J, Moser B, Mildner M, Ankersmit HJ. Toxicological testing of allogeneic secretome derived from peripheral mononuclear cells (APOSEC): a novel cell-free therapeutic agent in skin disease. Sci Rep 2019; 9:5598. [PMID: 30944367 PMCID: PMC6447581 DOI: 10.1038/s41598-019-42057-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 03/19/2019] [Indexed: 12/19/2022] Open
Abstract
A cell-free approach using secretomes derived from stem cells or peripheral blood mononuclear cells is an active area of regenerative medicine that holds promise for therapies. Regulatory authorities classify these secretomes as biological medicinal products, and non- clinical safety assessment thus falls under the scope of ICH S6. A secretome of stressed peripheral blood mononuclear cells (APOSEC) was successfully tested in a toxicology program, supporting clinical use of the new drug candidate. Here, to allow for topical, dermal treatment of patients with diabetic foot ulcer, several non-clinical safety studies were performed. Acute toxicity (single dose) and neuropharmacological screening were tested intravenously in a rat model. Risk for skin sensitisation was tested in mice. A 4-week intravenous toxicity study in mice and a 4-week subcutaneous toxicity study in minipigs were conducted to cover the clinical setting and application in a rodent and a non-rodent model. Acute and repeated-dose toxicity studies show that APOSEC administered intravenously and subcutaneously does not involve major toxicities or signs of local intolerance at levels above the intended total human maximal dose of 3.3 U/kg/treatment, 200 U/wound/treatment, and 100 U/cm2/treatment. The non-clinical data support the safe topical use of APOSEC in skin diseases related to deficient wound healing.
Collapse
Affiliation(s)
- Silvio Wuschko
- Drug and Chemical Safety Research & Toxicology, Consultant, Alland, Austria
| | - Alfred Gugerell
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,Department of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | | | - Helmut Hofbauer
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Maria Laggner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | | | | | - Anja Peterbauer
- Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Susanne Suessner
- Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Svitlana Demyanets
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Jost Leuschner
- LPT - Laboratory of Pharmacology and Toxicology GmbH & Co KG, Hamburg, Germany
| | - Bernhard Moser
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Hendrik J Ankersmit
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria. .,FFG Projects "APOSEC" 852748 and 862068, Medical University Vienna, Vienna, Austria.
| |
Collapse
|
24
|
Different pro-angiogenic potential of γ-irradiated PBMC-derived secretome and its subfractions. Sci Rep 2018; 8:18016. [PMID: 30573762 PMCID: PMC6301954 DOI: 10.1038/s41598-018-36928-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/27/2018] [Indexed: 12/11/2022] Open
Abstract
Secretomes from various cell sources exert strong regenerative activities on numerous organs, including the skin. Although secretomes consist of many diverse components, a growing body of evidence suggests that small extracellular vesicles (EVs) account for their regenerative capacity. We previously demonstrated that the secretome of γ-irradiated peripheral blood mononuclear cells (PBMCs) exhibits wound healing capacity. Therefore, we sought to dissect the molecular composition of EVs present in the secretome and compared wound healing-related activities of these EVs to other subfractions of the secretome and the fully supplemented secretome (MNCaposec). Compared to EVs derived from non-irradiated PBMCs, γ-irradiation significantly increased the size and number and changed the composition of released EVs. Detailed characterization of the molecular components of EVs, i.e. miRNA, proteins, and lipids, derived from irradiated PBMCs revealed a strong association with regenerative processes. Reporter gene assays and aortic ring sprouting assays revealed diminished activity of the subfractions compared to MNCaposec. In addition, we showed that MNCaposec accelerated wound closure in a diabetic mouse model. Taken together, our results suggest that secretome-based wound healing represents a promising new therapeutic avenue, and strongly recommend using the complete secretome instead of purified subfractions, such as EVs, to exploit its full regenerative capacity.
Collapse
|
25
|
Otero-Ortega L, Laso-García F, Gómez-de Frutos M, Fuentes B, Diekhorst L, Díez-Tejedor E, Gutiérrez-Fernández M. Role of Exosomes as a Treatment and Potential Biomarker for Stroke. Transl Stroke Res 2018; 10:241-249. [PMID: 30105420 DOI: 10.1007/s12975-018-0654-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/16/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022]
Abstract
Approximately, 16 million strokes occur worldwide each year, causing 6 million deaths and considerable disability, implying an enormous social, individual health, and economic burden. Due to this high incidence, strategies to promote stroke recovery are urgently needed. Research into new therapeutic approaches for stroke has determined that intravenous administration of mesenchymal stem cells (MSCs) is a good strategy to improve recovery by amplifying mechanisms implicated in brain plasticity. Recent studies have demonstrated the efficacy of MSCs in stroke, with no need for them to reach the area of brain injury. Although the mechanisms by which they mediate restorative effects are still unknown, the evidence suggests that MSCs might use specialised communication by sending and receiving biological information included in elements called exosomes. Exosomes are nanosized extracellular vesicles released into physical environments, and they have recently been suggested to mediate restorative stem cell effects. Moreover, after stroke, exosomes can also be synthesised and released from brain cells, passing through the blood-brain barrier (BBB), and can be detected in peripheral blood or in cerebrospinal fluid. Thus, exosomes could possibly be biomarkers that reflect pathological progress and promote stroke recovery. This review discusses the translational aspects of MSC-derived exosomes and their various roles in brain repair and as circulating biomarkers in stroke.
Collapse
Affiliation(s)
- Laura Otero-Ortega
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Fernando Laso-García
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - MariCarmen Gómez-de Frutos
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Blanca Fuentes
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Luke Diekhorst
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Exuperio Díez-Tejedor
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - María Gutiérrez-Fernández
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain.
| |
Collapse
|
26
|
Chen J, Chopp M. Exosome Therapy for Stroke. Stroke 2018; 49:1083-1090. [PMID: 29669873 PMCID: PMC6028936 DOI: 10.1161/strokeaha.117.018292] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Jieli Chen
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (J.C., M.C.)
- Department of Geriatrics, Tianjin Medical University General Hospital, China (J.C.)
- Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, China (J.C.)
| | - Michael Chopp
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (J.C., M.C.)
- Department of Physics, Oakland University, Rochester, MI (M.C.)
| |
Collapse
|
27
|
Diabetic Ephrin-B2-Stimulated Peripheral Blood Mononuclear Cells Enhance Poststroke Recovery in Mice. Stem Cells Int 2018; 2018:2431567. [PMID: 29736174 PMCID: PMC5875038 DOI: 10.1155/2018/2431567] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 11/18/2022] Open
Abstract
Clinical trials of cell therapy in stroke favor autologous cell transplantation. To date, feasibility studies have used bone marrow-derived mononuclear cells, but harvesting bone marrow cells is invasive thus complicating bedside treatment. We investigated the therapeutic potential of peripheral blood-derived mononuclear cells (PB-MNC) harvested from diabetic patients and stimulated by ephrin-B2 (PB-MNC+) (500,000 cells), injected intravenously 18–24 hours after induced cerebral ischemia in mice. Infarct volume, neurological deficit, neurogenesis, angiogenesis, and inflammation were investigated as were the potential mechanisms of PB-MNC+ cells in poststroke neurorepair. At D3, infarct volume was reduced by 60% and 49% compared to unstimulated PB-MNC and PBS-treated mice, respectively. Compared to PBS, injection of PB-MNC+ increased cell proliferation in the peri-infarct area and the subventricular zone, decreased microglia/macrophage cell density, and upregulated TGF-β expression. At D14, microvessel density was decreased and functional recovery was enhanced compared to PBS-treated mice, whereas plasma levels of BDNF, a major regulator of neuroplasticity, were increased in mice treated with PB-MNC+ compared to the other two groups. Cell transcriptional analysis showed that ephrin-B2 induced phenotype switching of PB-MNC by upregulating genes controlling cell proliferation, inflammation, and angiogenesis, as confirmed by adhesion and Matrigel assays. Conclusions. This feasibility study suggests that PB-MNC+ transplantation poststroke could be a promising approach but warrants further investigation. If confirmed, this rapid, noninvasive bedside cell therapy strategy could be applied to stroke patients at the acute phase.
Collapse
|
28
|
Haque N, Abdullah BJJ, Kasim NHA. Secretome: Pharmaceuticals for Cell-Free Regenerative Therapy. STEM CELL DRUGS - A NEW GENERATION OF BIOPHARMACEUTICALS 2018. [DOI: 10.1007/978-3-319-99328-7_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
29
|
Safety and tolerability of topically administered autologous, apoptotic PBMC secretome (APOSEC) in dermal wounds: a randomized Phase 1 trial (MARSYAS I). Sci Rep 2017; 7:6216. [PMID: 28740204 PMCID: PMC5524970 DOI: 10.1038/s41598-017-06223-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/20/2017] [Indexed: 12/26/2022] Open
Abstract
Developing effective therapies against chronic wound healing deficiencies is a global priority. Thus we evaluated the safety of two different doses of topically administered autologous APOSEC, the secretome of apoptotic peripheral blood mononuclear cells (PBMCs), in healthy male volunteers with artificial dermal wounds. Ten healthy men were enrolled in a single-center, randomized, double-blinded, placebo-controlled phase 1 trial. Two artificial wounds at the upper arm were generated using a 4-mm punch biopsy. Each participant was treated with both topically applied APOSEC and placebo in NuGel for 7 consecutive days. The volunteers were randomized into two groups: a low-dose group (A) receiving the supernatant of 12.5 × 106 PBMCs and a high-dose group (B) receiving an equivalent of 25 × 106 PBMCs resuspended in NuGel Hydrogel. Irradiated medium served as placebo. The primary outcome was the tolerability of the topical application of APOSEC. All adverse events were recorded until 17 days after the biopsy. Local tolerability assessment was measured on a 4-point scale. Secondary outcomes were wound closure and epithelization at day 7. No therapy-related serious adverse events occurred in any of the participants, and both low- and high-dose treatments were well tolerated. Wound closure was not affected by APOSEC therapy.
Collapse
|
30
|
Haque N, Kasim NHA, Kassim NLA, Rahman MT. Autologous serum supplement favours in vitro regenerative paracrine factors synthesis. Cell Prolif 2017; 50. [PMID: 28682474 DOI: 10.1111/cpr.12354] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 04/28/2017] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES Foetal bovine serum (FBS) is often the serum supplement of choice for in vitro human cell culture. This study compares the effect of FBS and autologous human serum (AuHS) supplement in human peripheral blood mononuclear cell (PBMC) culture to prepare secretome. MATERIALS AND METHODS The PBMC (n = 7) were cultured either in RPMI-1640 containing L-glutamine and 50 units/ml Penicillin-Streptomycin (BM) or in BM with either AuHS or FBS. Viability, proliferation and differentiation of PBMC were evaluated. Paracrine factors present in the secretomes (n = 6) were analysed using ProcartaPlex Human Cytokine panel (17 plex). Ingenuity Pathway Analysis (IPA) was performed to predict activation or inhibition of biological functions related to tissue regeneration. RESULTS The viability of PBMC that were cultured with FBS supplement was significantly reduced at 96 h compared to those at 0 and 24 h (P < .05). While the reduction of the viability of PBMC that were cultured with AuHS supplement was not significantly different compared to those at 0 and 24 h. The FBS secretomes prepared at 24 h was found to contain significantly higher amount of EGF (P < .05) compared to that in AuHS or BM secretome. The AuHS secretomes contained significantly higher amount of HGF at 24 (P < .05) and 96 h (P < .01), and VEGF-A at 24 h (P < .05) compared to those in the FBS secretomes. SDF-1 was not detected in the FBS secretomes prepared at either 24 or 96 hours. Double immunocytochemical staining revealed a marked increase in co-localization of SDF-1 and its receptor in PBMC that were cultured with AuHS supplement compared to that cultured with FBS supplement. CONCLUSION In secretome preparation, AuHS supplement favours synthesis of paracrine factors that are needed for regenerative therapy.
Collapse
Affiliation(s)
- Nazmul Haque
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.,Regenerative Dentistry Research Group, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.,Regenerative Dentistry Research Group, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Lide Abu Kassim
- Faculty of Education, International Islamic University Malaysia, Kuala Lumpur, Malaysia
| | | |
Collapse
|
31
|
Abstract
For almost two decades, cell-based therapies have been tested in modern regenerative medicine to either replace or regenerate human cells, tissues, or organs and restore normal function. Secreted paracrine factors are increasingly accepted to exert beneficial biological effects that promote tissue regeneration. These factors are called the cell secretome and include a variety of proteins, lipids, microRNAs, and extracellular vesicles, such as exosomes and microparticles. The stem cell secretome has most commonly been investigated in pre-clinical settings. However, a growing body of evidence indicates that other cell types, such as peripheral blood mononuclear cells (PBMCs), are capable of releasing significant amounts of biologically active paracrine factors that exert beneficial regenerative effects. The apoptotic PBMC secretome has been successfully used pre-clinically for the treatment of acute myocardial infarction, chronic heart failure, spinal cord injury, stroke, and wound healing. In this review we describe the benefits of choosing PBMCs instead of stem cells in regenerative medicine and characterize the factors released from apoptotic PBMCs. We also discuss pre-clinical studies with apoptotic cell-based therapies and regulatory issues that have to be considered when conducting clinical trials using cell secretome-based products. This should allow the reader to envision PBMC secretome-based therapies as alternatives to all other forms of cell-based therapies.
Collapse
Affiliation(s)
- Lucian Beer
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Mariann Gyöngyösi
- Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Medical University of Vienna, Vienna, Austria.
- Head FFG Project 852748 "APOSEC", FOLAB Surgery, Medical University of Vienna, Vienna, Austria.
- Department of Thoracic Surgery, Medical University Vienna, Vienna, Austria.
| |
Collapse
|
32
|
Kasiri MM, Beer L, Nemec L, Gruber F, Pietkiewicz S, Haider T, Simader EM, Traxler D, Schweiger T, Janik S, Taghavi S, Gabriel C, Mildner M, Ankersmit HJ. Dying blood mononuclear cell secretome exerts antimicrobial activity. Eur J Clin Invest 2016; 46:853-63. [PMID: 27513763 PMCID: PMC5113772 DOI: 10.1111/eci.12667] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 08/09/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Several activities are attributed to antimicrobial peptides (AMPs), including bacterial killing, leucocyte recruitment and angiogenesis. Despite promises of advanced cellular therapies for treatment of diabetic foot ulcer, it is currently accepted that paracrine factors rather than cellular components are causative for the observed effects. Whether AMPs are present in the mononuclear cell (MNC) secretome (MNC-sec) of white blood cells that are beneficial in experimental wound healing is not known. MATERIALS AND METHODS Antimicrobial activity of the secretomes of nonirradiated (MNC-sec) and γ-irradiated MNCs (MNC-sec rad) was analysed by microdilution assay. AMPs were determined by quantitative real-time PCR (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). Whether human MNC-sec rad causes AMP secretion in vivo was examined in an experimental rat model. Image flow cytometry was used to determine the type of cell death induced in MNCs after exposure to γ-radiation. RESULTS The antimicrobial activity assay revealed a bactericidal activity of MNC-sec rad and to a lesser degree also of MNC-sec. Image flow cytometry showed that γ-irradiation of MNCs induced early apoptosis followed mainly by necroptosis. RT-PCR and ELISA revealed a high abundance of different AMPs in the secretome of MNCs. In addition, human MNC-sec elicited an increase in de novo endogenous AMP production in rats in vivo. CONCLUSION We provide evidence that the secretome of MNCs has direct and indirect positive effects on the immune defence system, including augmentation of antibacterial properties. Our data further suggest that necroptosis could play a key role for the release of paracrine factors and the therapeutic action of MNC-sec rad.
Collapse
Affiliation(s)
- Mohammad Mahdi Kasiri
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria
| | - Lucian Beer
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria.,Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Lucas Nemec
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Florian Gruber
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
| | - Sabine Pietkiewicz
- Translational Inflammation Research, Otto von Guericke University, Magdeburg, Germany
| | - Thomas Haider
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria
| | - Elisabeth Maria Simader
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria
| | - Denise Traxler
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria
| | - Thomas Schweiger
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Stefan Janik
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria
| | - Shahrokh Taghavi
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| | - Hendrik Jan Ankersmit
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria. .,Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria. .,Head FFG Project 852748 'APOSEC', Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
33
|
Analysis of the Secretome of Apoptotic Peripheral Blood Mononuclear Cells: Impact of Released Proteins and Exosomes for Tissue Regeneration. Sci Rep 2015; 5:16662. [PMID: 26567861 PMCID: PMC4645175 DOI: 10.1038/srep16662] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/19/2015] [Indexed: 12/12/2022] Open
Abstract
We previously showed that, when peripheral blood mononuclear cells (PBMCs) were stressed with ionizing radiation, they released paracrine factors that showed regenerative capacity in vitro and in vivo. This study aimed to characterize the secretome of PBMCs and to investigate its biologically active components in vitro and vivo. Bioinformatics analysis revealed that irradiated PBMCs differentially expressed genes that encoded secreted proteins. These genes were primarily involved in (a) pro-angiogenic and regenerative pathways and (b) the generation of oxidized phospholipids with known pro-angiogenic and inflammation-modulating properties. Subsequently, in vitro assays showed that the exosome and protein fractions of irradiated and non-irradiated PBMC secretome were the major biological components that enhanced cell mobility; conversely, secreted lipids and microparticles had no effects. We tested a viral-cleared PBMC secretome, prepared according to good manufacturing practice (GMP), in a porcine model of closed chest, acute myocardial infarction. We found that the potency for preventing ventricular remodeling was similar with the GMP-compliant and experimentally-prepared PBMC secretomes. Our results indicate that irradiation modulates the release of proteins, lipid-mediators and extracellular vesicles from human PBMCs. In addition our findings implicate the use of secretome fractions as valuable material for the development of cell-free therapies in regenerative medicine.
Collapse
|