Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 387
Filter
1.
J Neuroimmune Pharmacol ; 19(1): 52, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39382610

ABSTRACT

Cirrhotic patients may show minimal hepatic encephalopathy (MHE), with mild cognitive impairment and motor deficits. Hyperammonemia and inflammation are the main contributors to the cognitive and motor alterations of MHE. Hyperammonemic rats reproduce these alterations. There are no specific treatments for the neurological alterations of MHE. Extracellular vesicles from mesenchymal stem cells (MSC-EVs) are promising to treat inflammatory and immune diseases. We aimed to assess whether treatment of hyperammonemic rats with MSC-EVs reduced neuroinflammation in cerebellum and restored motor coordination and to study the mechanisms involved. The effects of MSC-EVs were studied in vivo by intravenous injection to hyperammonemic rats and ex vivo in cerebellar slices. Motor coordination was analyzed using the beam walking test. Effects on neuroinflammation were assessed by immunohistochemistry, immunofluorescence and Western blot. Injection of MSC-EVs reduced microglia and astrocytes activation in cerebellum and restored motor coordination in hyperammonemic rats. Ex vivo experiments show that MSC-EVs normalize pro-inflammatory factors, including TNFα, NF-kB activation and the activation of two key pathways leading to motor incoordination (TNFR1-NF-kB-glutaminase-GAT3 and TNFR1-CCL2-BDNF-TrkB-KCC2). TGFß in the EVs was necessary for these beneficial effects. MSC-EVs treatment reverse neuroinflammation in the cerebellum of hyperammonemic rats and the underlying mechanisms leading to motor incoordination. Therapy with MSC-EVs may be useful to improve motor function in patients with MHE.


Subject(s)
Extracellular Vesicles , Hyperammonemia , Mesenchymal Stem Cells , Rats, Wistar , Animals , Extracellular Vesicles/transplantation , Extracellular Vesicles/metabolism , Hyperammonemia/therapy , Hyperammonemia/metabolism , Rats , Male , Mesenchymal Stem Cells/metabolism , Neuroinflammatory Diseases/therapy , Cerebellum/metabolism , Hepatic Encephalopathy/therapy , Hepatic Encephalopathy/metabolism
2.
Sci Rep ; 14(1): 23538, 2024 10 09.
Article in English | MEDLINE | ID: mdl-39384602

ABSTRACT

In adult mammals, limb regeneration is limited by the absence of blastemal cells (BCs) and the lack of the regenerative signaling cascade. The utilization of transgenic cells circumvents the limitations associated with the absence of BCs. In a previous investigation, we successfully regenerated mouse phalanx amputations using blastema-like cells (BlCs) generated from bone marrow-derived mesenchymal stem cells (mBMSCs) overexpressing Msx1 and Msx2 genes. Recently, extracellular vesicles (EVs) have emerged as potent biological tools, offering a promising alternative to manipulated cells for clinical applications. This research focuses on utilizing BlCs-derived extracellular vesicles (BlCs-EVs) for regenerating mouse digit tips. The BlCs were cultured and expanded, and then EVs were isolated via ultracentrifugation. The size, morphology, and CD81 marker expression of the EVs were confirmed through Dynamic Light Scattering (DLS), Scanning Electron Microscope (SEM), and Western Blot (WB) analyses. Additionally, WB analysis demonstrated the presence of MSX1, MSX2, FGF8, and BMP4 proteins. The uptake of EVs by mBMSCs was shown through immunostaining. Effects on cell proliferation, migration, and osteogenic activity post-treatment with BlCs-EVs were assessed through MTT assay, scratch assay, and Real-time PCR. The regenerative potential of BlCs-EVs was evaluated in a mouse digit tip amputation model using histological assessments. Results indicated that BlCs-EVs enhanced several abilities of mBMSCs, such as migration, proliferation, and osteogenesis in vitro. Notably, BlCs-EVs significantly improved digit tip regeneration in mice, promoting the formation of new bone and nails, which was absent in control groups. In summary, BlCs-EVs are promising tools for digit tip regeneration, avoiding the ethical concerns associated with using genetically modified cells.


Subject(s)
Extracellular Vesicles , MSX1 Transcription Factor , Mesenchymal Stem Cells , Regeneration , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Mice , MSX1 Transcription Factor/metabolism , MSX1 Transcription Factor/genetics , Cell Proliferation , Osteogenesis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Disease Models, Animal , Cell Movement , Fibroblast Growth Factor 8/metabolism , Fibroblast Growth Factor 8/genetics
3.
Stem Cell Res Ther ; 15(1): 300, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39272156

ABSTRACT

BACKGROUND: Emerging evidence has highlighted the therapeutic potential of human umbilical cord mesenchymal stem cells (UC-MSCs) in chemotherapy-induced premature ovarian failure (POF). This study was designed to investigate the appropriate timing and molecular mechanism of UC-MSCs treatment for chemotherapy-induced POF. METHODS: Ovarian structure and function of mice were assessed every 3 days after injections with cyclophosphamide (CTX) and busulfan (BUS). UC-MSCs and UC-MSCs-derived extracellular vesicles (EVs) were infused into mice via the tail vein, respectively. Ovarian function was analyzed by follicle counts, the serum levels of hormones and ovarian morphology. The apoptosis and proliferation of ovarian granulosa cells were analyzed in vitro and in vivo. Label-free quantitative proteomics was used to detect the differentially expressed proteins in UC-MSC-derived EVs. RESULTS: After CTX/BUS injection, we observed that the ovarian function of POF mice was significantly deteriorated on day 9 after CTX/BUS infusion. TUNEL assay indicated that the number of apoptotic cells in the ovaries of POF mice was significantly higher than that in normal mice on day 3 after CTX/BUS injection. Transplantation of UC-MSCs on day 6 after CTX/BUS injection significantly improved ovarian function, enhanced proliferation and inhibited apoptosis of ovarian granulosa cells, whereas the therapeutic effect of UC-MSCs transplantation decreased on day 9, or day 12 after CTX/BUS injection. Moreover, EVs derived from UC-MSCs exerted similar therapeutic effects on POF. UC-MSCs-derived EVs could activate the PI3K/AKT signaling pathway and reduce ovarian granulosa cell apoptosis. Quantitative proteomics analysis revealed that clusterin (CLU) was highly expressed in the EVs of UC-MSCs. The supplementation of CLU proteins prevented ovarian granulosa cells from chemotherapy-induced apoptosis. Further mechanistic analysis showed that CLU-knockdown blocked the PI3K/AKT signaling and reversed the protective effects of UC-MSCs-derived EVs. CONCLUSIONS: Administration of UC-MSCs and UC-MSCs-derived EVs on day 6 of CTX/BUS injection could effectively improve the ovarian function of POF mice. UC-MSCs-derived EVs carrying CLU promoted proliferation and inhibited apoptosis of ovarian granulosa cells through activating the PI3K/AKT pathway. This study identifies a previously unrecognized molecular mechanism of UC-MSCs-mediated protective effects on POF, which pave the way for the use of cell-free therapeutic approach for POF.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Phosphatidylinositol 3-Kinases , Primary Ovarian Insufficiency , Proto-Oncogene Proteins c-akt , Signal Transduction , Umbilical Cord , Female , Animals , Primary Ovarian Insufficiency/therapy , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/chemically induced , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Proto-Oncogene Proteins c-akt/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Umbilical Cord/cytology , Clusterin/metabolism , Apoptosis , Mesenchymal Stem Cell Transplantation/methods , Ovary/metabolism , Granulosa Cells/metabolism , Cell Proliferation , Busulfan/pharmacology
4.
Front Biosci (Landmark Ed) ; 29(9): 340, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39344329

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) poses a significant public health challenge, increasingly affecting patients' finances, mental health, and functional abilities as the global population ages. Stem cell-derived extracellular vesicles (SC-EVs) have emerged as a promising cell-free therapeutic approach for AD, although their precise mechanisms remain unclear. This meta-analysis aims to evaluate the effectiveness of SC-EVs in treating AD. METHODS: We systematically searched PubMed, EMBASE, and Web of Science databases up to December 31, 2023, identifying studies investigating SC-EVs therapy in AD rodent models. Outcome measures included Morris water maze and Y maze tests, ß-amyloid pathology, and inflammatory markers. Statistical analyses utilized Stata 15.1 and R software. RESULTS: This meta-analysis of 16 studies (2017-2023, 314 animals) demonstrates significant efficacy of SC-EVs therapy in AD models. Pooled analyses demonstrated that SC-EVs therapy significantly increased the learning function as measured by Morris water maze tests (MWM) by -1.83 (95% CI = -2.51 to -1.15, p < 0.0001), Y maze test by 1.66 (95% CI = 1.03 to 2.28, p < 0.0001), decreased Aß plaques in the hippocampal by -2.10 (95% CI = -2.96 to -1.23, p < 0.0001), and proinflammatory cytokines Tumor necrosis factor alpha (TNFα) by -2.61 (95% CI = -4.87 to -0.35, p < 0.05), Interleukin-1 beta (IL-1ß) by -2.37 (95% CI = -3.68 to -1.05, p < 0.001). CONCLUSIONS: SC-EVs therapy shows promise in enhancing cognitive function and mitigating AD progression in preclinical models. Future research should focus on standardizing methodologies and comparing SC-EVs isolation techniques and dosing strategies to facilitate clinical translation.


Subject(s)
Alzheimer Disease , Extracellular Vesicles , Alzheimer Disease/therapy , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Animals , Humans , Disease Models, Animal , Stem Cells/cytology , Maze Learning , Amyloid beta-Peptides/metabolism
5.
Int Immunopharmacol ; 142(Pt A): 113104, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39270344

ABSTRACT

Mitochondrial dysfunction has been identified as a trigger for cellular autophagy dysfunction and programmed cell death. Emerging studies have revealed that, in pathological contexts, intercellular transfer of mitochondria takes place, facilitating the restoration of mitochondrial function, energy metabolism, and immune homeostasis. Extracellular vesicles, membranous structures released by cells, exhibit reduced immunogenicity and enhanced stability during the transfer of mitochondria. Thus, this review provides a concise overview of mitochondrial dysfunction related diseases and the mechanism of mitochondrial dysfunction in diseases progression, and the composition and functions of the extracellular vesicles, along with elucidating the principal mechanisms underlying intercellular mitochondrial transfer. In this article, we will focus on the advancements in both animal models and clinical trials concerning the therapeutic efficacy of extracellular vesicle-mediated mitochondrial transplantation across various systemic diseases in neurodegenerative diseases and cardiovascular diseases. Additionally, the review delves into the multifaceted roles of extracellular vesicle-transplanted mitochondria, encompassing anti-inflammatory actions, promotion of tissue repair, enhancement of cellular function, and modulation of metabolic and immune homeostasis within diverse pathological contexts, aiming to provide novel perspectives for extracellular vesicle transplantation of mitochondria in the treatment of various diseases.


Subject(s)
Exosomes , Mitochondria , Humans , Animals , Exosomes/metabolism , Exosomes/transplantation , Mitochondria/metabolism , Mitochondria/transplantation , Neurodegenerative Diseases/therapy , Neurodegenerative Diseases/immunology , Cardiovascular Diseases/therapy , Cardiovascular Diseases/metabolism , Extracellular Vesicles/transplantation , Extracellular Vesicles/metabolism , Mitochondrial Diseases/therapy , Mitochondrial Diseases/immunology , Mitochondrial Diseases/metabolism
6.
Biochim Biophys Acta Rev Cancer ; 1879(5): 189177, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39218403

ABSTRACT

Cell therapy has emerged as one of the most promising approaches to treating disease in recent decades. The application of stem cells in anti-tumor therapy is determined by their varying capacity for proliferation, migration, and differentiation. These capacities are derived from different sources. The use of stem cell carriers in cancer treatment is justified by the following three reasons: (I) shield therapeutic agents from swift biological deterioration; (II) reduce systemic side effects; and (III) increase local therapeutic levels since stem cells have an innate ability to target tumors. The quantity of stem cells confined to the tumor microenvironment determines this system's anti-tumor activity. Nevertheless, there are limitations to the use of different types of stem cells. When immune cells are used in cell therapy, it may lead to cytokine storms and improper reactions to self-antigens. Furthermore, the use of stem cells may result in cancer. Additionally, after an intravenous injection, cells could not migrate to the injury location. Exosomes derived from different cells were thus proposed as possible therapeutic options. Exosomes are becoming more and more well-liked because of their small size, biocompatibility, and simplicity in storage and separation. A number of investigations have shown that adding various medications and microRNAs to exosomes may enhance their therapeutic effectiveness. Thus, it is essential to evaluate studies looking into the therapeutic effectiveness of encapsulated exosomes. In this review, we looked at studies on encapsulated exosomes' use in regenerative medicine and the treatment of cancer. The results imply that the therapeutic potential increases when encapsulated exosomes are used rather than intact exosomes. Therefore, in order to optimize the effectiveness of the treatment, it is advised to implement this technique in accordance with the kind of therapy.


Subject(s)
Extracellular Vesicles , Hydrogels , Mesenchymal Stem Cells , Neoplasms , Humans , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/pathology , Mesenchymal Stem Cells/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Animals , Exosomes/metabolism , Tumor Microenvironment , Mesenchymal Stem Cell Transplantation/methods
7.
Circ Res ; 135(8): 877-885, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39325847

ABSTRACT

All Food and Drug Administration-approved noncoding RNA (ncRNA) drugs (n≈20) target known disease-causing molecular pathways by mechanisms such as antisense. In a fortuitous evolution of work on regenerative medicine, my coworkers and I inverted the RNA drug discovery process: first we identified natural disease-modifying ncRNAs, then used them as templates for new synthetic RNA drugs. Mechanism was probed only after bioactivity had been demonstrated. The journey began with the development of cardiosphere-derived cells (CDCs) for cardiac regeneration. While testing CDCs in a first-in-human trial, we discovered they worked indirectly: ncRNAs within CDC-secreted extracellular vesicles mediate the therapeutic benefits. The vast majority of such ncRNAs are fragments of unknown function. We chose several abundant ncRNA species from CDC-secreted extracellular vesicles, synthesized and screened each of them in vitro and in vivo. Those with exceptional disease-modifying bioactivity inspired new chemical entities that conform to the structural conventions of the Food and Drug Administration-approved ncRNA armamentarium. This discovery arc-Cell-Derived RNA from Extracellular vesicles for bioinspired Drug develOpment, or CREDO-has yielded various promising lead compounds, each of which works via a unique, and often novel, mechanism. The process relies on emergent insights to shape therapeutic development. The initial focus of our inquiry-CDCs-are now themselves in phase 3 testing for Duchenne muscular dystrophy and its associated cardiomyopathy. But the intravenous delivery strategy and the repetitive dosing protocol for CDCs, which have proven key to clinical success, both arose from systematic mechanistic inquiry. Meanwhile, emergent insights have led to multiple cell-free therapeutic candidates: CDC-secreted extracellular vesicles are in preclinical development for ventricular arrhythmias, while the CREDO-conceived RNA drugs are in translation for diseases ranging from myocarditis to scleroderma.


Subject(s)
Extracellular Vesicles , RNA, Untranslated , Regenerative Medicine , Humans , Regenerative Medicine/methods , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , RNA, Untranslated/metabolism , RNA, Untranslated/genetics , Cell- and Tissue-Based Therapy/methods , Drug Discovery/methods , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects
8.
Z Orthop Unfall ; 162(5): 533-548, 2024 Oct.
Article in German | MEDLINE | ID: mdl-39321815

ABSTRACT

The principle of regenerative medicine in the treatment of osteoarthritis pursues a functional restoration of cartilage tissue instead of just repairing cartilage defects. The use of blood products is intended to inhibit chronic inflammatory processes and promote tissue regeneration. Intraarticular injection of autologous platelet-rich plasma (PRP) is a prominent procedure. Clinical evidence supports PRP injection over hyaluronic acid or glucocorticoid injection. Comparability of studies is difficult due to missing standardisation of production procedures, dosing and donor variability. In particular, whether presence of residual leukocytes is required or should be avoided is an open debate. In contrast, stem cell therapies in osteoarthritis therapy are often based on mesenchymal stem cells (MSC) from adipose tissue or bone marrow aspirate. Different sources of MSC might render the cells more suitable for application in a given context. Nevertheless, it became evident that their secretome rather than the cells themselves are responsible for observed regenerative processes. Research on the mechanisms of action have focused on growth factors. However, an overlooked component of blood products called extracellular vesicles (EV) came to the center of attention, which are also released by MSC as intercellular signal carriers. EV cargo molecules such as miRNAs open up new dimensions in the investigation and explanation of clinically observed anti-inflammatory and regenerative effects.


Subject(s)
Mesenchymal Stem Cell Transplantation , Osteoarthritis , Platelet-Rich Plasma , Humans , Osteoarthritis/therapy , Mesenchymal Stem Cell Transplantation/methods , Injections, Intra-Articular , Extracellular Vesicles/transplantation
9.
Biomed Pharmacother ; 178: 117256, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39111081

ABSTRACT

INTRODUCTION: Insufficient supply of cardiac grafts represents a severe obstacle in heart transplantation. Donation after Circulatory Death (DCD), in addition to conventional donation after brain death, is one promising option to overcome the organ shortage. However, DCD organs undergo an inevitable more extended period of warm unprotected ischemia between circulatory arrest and graft procurement. Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) have shown remarkable protective effects against ischemia-reperfusion injury. Thus, we aimed to enhance grafts preservation from DCD donors, through treatment with MSC-EVs. METHODS: Female pigs were euthanized by barbiturate overdose and after 20 min of a flat EKG, the chest was opened, the heart harvested and subsequently connected to an extracorporeal perfusion machine. MSC-EVs, isolated by ion exchange chromatography, were added to the perfusion solution (1×1011 particles) and the heart was perfused for 2 h. Then, heart tissue biopsies were taken to assess histological changes, mitochondrial morphology, antioxidant enzyme activity and inflammation mediators' expression. Biochemical parameters of myocardial viability were assessed in the perfusate. RESULTS: The treatment with MSC-EVs significantly prevented mitochondria swelling, mitochondrial cristae loss and oxidative stress in cardiac tissue. The protective effect of MSC-EVs was confirmed by the delayed increase of the cardiac-specific enzymes CK and TnC in the perfusate and the reduction of caspase-3+ cells in tissue sections. CONCLUSION: MSC-EVs improve graft quality by preserving the mitochondrial ultrastructure protecting the myocardium against oxidative stress, reducing apoptosis of cardiac cells and preventing the increase of pro-inflammatory cytokines.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Mesenchymal Stem Cells/metabolism , Female , Swine , Oxidative Stress , Heart Transplantation/methods , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/pathology , Disease Models, Animal , Myocardium/pathology , Myocardium/metabolism , Reperfusion Injury/prevention & control , Reperfusion Injury/pathology
10.
ACS Biomater Sci Eng ; 10(9): 5784-5795, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39164977

ABSTRACT

Extracellular vesicles derived from mesenchymal stem cells (MSCs-EVs) have great potential for bone remodeling and anti-inflammatory therapy. For the repair and reconstruction of inflammatory jawbone defects caused by periapical periodontitis, bone meal filling after debridement is commonly used in the clinic. However, this treatment has disadvantages such as large individual differences and the need for surgical operation. Therefore, it is of great significance to search for other bioactive substances that can promote jawbone regeneration in periapical periodontitis. Herein, it is found that CT results showed that local injection of human umbilical cord mesenchymal stem cells-derived extracellular vesicles (HUC-MSCs-EVs) and bone meal filling into the alveolar bone defect area could promote bone tissue regeneration using a rat model of a jawbone defect in periapical periodontitis. Histologically, the new periodontal tissue in the bone defect area was thicker, and the number of blood vessels was higher by local injection of HUC-MSCs-EVs, and fewer inflammatory cells and osteoclasts were formed compared to bone meal filling. In vitro, HUC-MSCs-EVs can be internalized by rat bone marrow mesenchymal stem cells (BMSCs), enhancing the ability for proliferation and migration of BMSCs. Additionally, 20 µg/mL HUC-MSCs-EVs can facilitate the expression of osteogenic genes and proteins including runt-related transcription factor 2 (RUNX2), alkaline phosphatase (ALP), and osteopontin (OPN). In summary, in vivo and in vitro experiments showed that HUC-MSCs-EVs can promote bone regeneration in periapical periodontitis, and the effect of tissue regeneration is better than that of traditional bone meal treatment. Therefore, local injection of HUC-MSCs-EVs may be an effective method to promote jawbone regeneration in periapical periodontitis.


Subject(s)
Bone Regeneration , Extracellular Vesicles , Mesenchymal Stem Cells , Periapical Periodontitis , Umbilical Cord , Animals , Mesenchymal Stem Cells/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Humans , Periapical Periodontitis/therapy , Periapical Periodontitis/metabolism , Periapical Periodontitis/pathology , Bone Regeneration/physiology , Rats , Umbilical Cord/cytology , Male , Rats, Sprague-Dawley , Cell Proliferation , Mesenchymal Stem Cell Transplantation/methods , Osteopontin/metabolism , Osteogenesis
11.
Aging Dis ; 15(5): 2191-2204, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39191396

ABSTRACT

Ischemic stroke represents a significant global health challenge, often resulting in death or long-term disability, particularly among the elderly, where advancing age stands as the most unmodifiable risk factor. Arising from the blockage of a brain-feeding artery, the only therapies available to date aim at removing the blood clot to restore cerebral blood flow and rescue neuronal cells from death. The prevailing treatment approach involves thrombolysis by administration of recombinant tissue plasminogen activator (tPA), albeit with a critical time constraint. Timely intervention is imperative, given that delayed thrombolysis increases tPA leakage into the brain parenchyma, causing harmful effects. Strategies to preserve tPA's vascular benefits while shielding brain cells from its toxicity have been explored. Notably, administering neuroserpin (Ns), a brain-specific tPA inhibitor, represents one such approach. Following ischemic stroke, Ns levels rise and correlate with favorable post-stroke outcomes. Studies in rodent models of focal cerebral ischemia have demonstrated the beneficial effects of Ns administration. Ns treatment maintains blood-brain barrier (BBB) integrity, reducing stroke volume. Conversely, Ns-deficient animals exhibit larger stroke injury, increased BBB permeability and enhanced microglia activation. Furthermore, Ns administration extends the therapeutic window for tPA intervention, underscoring its potential in stroke management. Remarkably, our investigation reveals the presence of Ns within extracellular vesicles (EVs), small membrane-surrounded particles released by all cells and critical for intercellular communication. EVs influence disease outcome following stroke through cargo transfer between cells. Clarifying the role of EVs containing NS could open up urgently needed novel therapeutic approaches to improve post-ischemic stroke outcome.


Subject(s)
Extracellular Vesicles , Ischemic Stroke , Neuroserpin , Serpins , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Animals , Humans , Serpins/metabolism , Serpins/therapeutic use , Neuropeptides/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Neuroprotection/drug effects , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Tissue Plasminogen Activator/metabolism , Tissue Plasminogen Activator/therapeutic use
12.
Am J Physiol Cell Physiol ; 327(4): C929-C945, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39099421

ABSTRACT

In the context of improving the efficacy of autologous fat grafts (AFGs) in reconstructive surgery, this study delineates the novel use of adipose-derived mesenchymal stem cells (ADSCs) and their extracellular vesicles (EVs) as vehicles for delivering delta-like ligand 4 (DLL4) siRNA. The aim was to inhibit DLL4, a gene identified through transcriptome analysis as a critical player in the vascular endothelial cells of AFG tissues, thereby negatively affecting endothelial cell functions and graft survival through the Notch signaling pathway. By engineering ADSC EVs to carry DLL4 siRNA (ADSC EVs-siDLL4), the research demonstrated a marked improvement in endothelial cell proliferation, migration, and lumen formation, and enhanced angiogenesis in vivo, leading to a significant increase in the survival rate of AFGs. This approach presents a significant advancement in the field of tissue engineering and regenerative medicine, offering a potential method to overcome the limitations of current fat grafting techniques.NEW & NOTEWORTHY This study introduces a groundbreaking method for enhancing autologous fat graft survival using adipose-derived stem cell extracellular vesicles (ADSC EVs) to deliver DLL4 siRNA. By targeting the delta-like ligand 4 (DLL4) gene, crucial in endothelial cell dynamics, this innovative approach significantly improves endothelial cell functions and angiogenesis, marking a substantial advancement in tissue engineering and regenerative medicine.


Subject(s)
Adaptor Proteins, Signal Transducing , Adipose Tissue , Calcium-Binding Proteins , Endothelial Cells , Extracellular Vesicles , Mesenchymal Stem Cells , RNA, Small Interfering , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Extracellular Vesicles/genetics , Animals , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Endothelial Cells/metabolism , Adipose Tissue/metabolism , Adipose Tissue/cytology , Mesenchymal Stem Cells/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , Neovascularization, Physiologic , Cell Proliferation , Mice , Signal Transduction , Graft Survival/physiology , Cells, Cultured , Cell Movement
13.
Respir Res ; 25(1): 303, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112999

ABSTRACT

BACKGROUND: Acute lung injury (ALI) following pneumonia involves uncontrolled inflammation and tissue injury, leading to high mortality. We previously confirmed the significantly increased cargo content and extracellular vesicle (EV) production in thrombin-preconditioned human mesenchymal stromal cells (thMSCs) compared to those in naïve and other preconditioning methods. This study aimed to investigate the therapeutic efficacy of EVs derived from thMSCs in protecting against inflammation and tissue injury in an Escherichia coli (E. coli)-induced ALI mouse model. METHODS: In vitro, RAW 264.7 cells were stimulated with 0.1 µg/mL liposaccharides (LPS) for 1 h, then were treated with either PBS (LPS Ctrl) or 5 × 107 particles of thMSC-EVs (LPS + thMSC-EVs) for 24 h. Cells and media were harvested for flow cytometry and ELISA. In vivo, ICR mice were anesthetized, intubated, administered 2 × 107 CFU/100 µl of E. coli. 50 min after, mice were then either administered 50 µL saline (ECS) or 1 × 109 particles/50 µL of thMSC-EVs (EME). Three days later, the therapeutic efficacy of thMSC-EVs was assessed using extracted lung tissue, bronchoalveolar lavage fluid (BALF), and in vivo computed tomography scans. One-way analysis of variance with post-hoc TUKEY test was used to compare the experimental groups statistically. RESULTS: In vitro, IL-1ß, CCL-2, and MMP-9 levels were significantly lower in the LPS + thMSC-EVs group than in the LPS Ctrl group. The percentages of M1 macrophages in the normal control, LPS Ctrl, and LPS + thMSC-EV groups were 12.5, 98.4, and 65.9%, respectively. In vivo, the EME group exhibited significantly lower histological scores for alveolar congestion, hemorrhage, wall thickening, and leukocyte infiltration than the ECS group. The wet-dry ratio for the lungs was significantly lower in the EME group than in the ECS group. The BALF levels of CCL2, TNF-a, and IL-6 were significantly lower in the EME group than in the ECS group. In vivo CT analysis revealed a significantly lower percentage of damaged lungs in the EME group than in the ECS group. CONCLUSION: Intratracheal thMSC-EVs administration significantly reduced E. coli-induced inflammation and lung tissue damage. Overall, these results suggest therapeutically enhanced thMSC-EVs as a novel promising therapeutic option for ARDS/ALI.


Subject(s)
Acute Lung Injury , Extracellular Vesicles , Mesenchymal Stem Cells , Mice, Inbred ICR , Thrombin , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Acute Lung Injury/metabolism , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Acute Lung Injury/therapy , Mice , Mesenchymal Stem Cells/metabolism , RAW 264.7 Cells , Thrombin/metabolism , Escherichia coli , Male , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli Infections/therapy , Treatment Outcome , Disease Models, Animal , Humans
14.
Sci Rep ; 14(1): 19273, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164295

ABSTRACT

Extracellular vesicles (EVs) from mesenchymal stem cells (MSCs), specifically those preconditioned with deferoxamine (DFO) in canine adipose tissue-derived MSCs (cAT-MSCs), were explored for treating autoimmune diseases. This study assessed the effects of DFO-preconditioned EVs (EVDFO) in an experimental autoimmune encephalomyelitis (EAE) mouse model. cAT-MSCs were treated with DFO for 48 h, after which EVs were isolated. EAE mice received intranasal EV or EVDFO treatments and were euthanized following histopathologic analysis; RNA and protein expression levels were measured. Histologically, EV and EVDFO groups showed a significant reduction in inflammatory cell infiltration and demyelination. Immunofluorescence revealed increased CD206 and Foxp3 expression, indicating elevated M2 macrophages and regulatory T (Treg) cells, particularly in the EVDFO group. Treg cells also notably increased in the spleen of EVDFO -treated mice. STAT3 and pSTAT3 proteins were upregulated in the EAE groups compared to the naïve group. However, following EV treatment, STAT3 expression decreased compared to the EAE group, whereas pSTAT3 expression was similar in both the EV and EAE groups. In conclusion, EVDFO treatment resulted in reduced STAT3 expression, suggesting its role in T cell regulation and the potential of EVDFO in modulating the STAT3 pathway for reducing inflammation more effectively than non-preconditioned EVs.


Subject(s)
Deferoxamine , Encephalomyelitis, Autoimmune, Experimental , Extracellular Vesicles , Inflammation , Mesenchymal Stem Cells , STAT3 Transcription Factor , T-Lymphocytes, Regulatory , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , STAT3 Transcription Factor/metabolism , Mice , Dogs , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Mesenchymal Stem Cells/metabolism , Inflammation/pathology , Female , Disease Models, Animal
15.
Cells ; 13(16)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39195224

ABSTRACT

Under vasculogenic conditioning, certain pro-inflammatory subsets within peripheral blood mononuclear cells (PBMCs) undergo phenotypic transformation into pro-regenerative types, such as vasculogenic endothelial progenitor cells, M2 macrophages, and regulatory T cells. These transformed cells are collectively termed regeneration-associated cells (RACs). In this study, we aimed to investigate the therapeutic efficacy of RAC-derived extracellular vesicles (RACev) compared with a vehicle-treated group in the context of renal ischemia-reperfusion injury (R-IRI). Human PBMCs were cultured with defined growth factor cocktails for seven days to harvest RACs. EV quantity and size were characterized by nanoparticle tracking analysis. Notably, the systemic injection of RACev significantly decreased serum creatinine and blood urine nitrogen at day three compared to the control group. Histologically, the treatment group showed less fibrosis in the cortex and medullary areas (p < 0.04 and p < 0.01) compared to the control group. The CD31 staining confirmed enhanced capillary densities in the treatment group compared to the control group (p < 0.003). These beneficial effects were accompanied by angiogenesis, anti-fibrosis, anti-inflammation, and anti-apoptosis RACev miR delivery to ischemic injury to control inflammatory, endothelial mesenchymal transition, and hypoxia pathways. In vivo bioluminescence analysis demonstrated a preferential accumulation of RACev in the IR-injured kidney. The systemic transplantation of RACev beneficially restored kidney function by protecting from tissue fibrosis and through anti-inflammation, angiogenesis, and anti-apoptosis miR delivery to the ischemic tissue.


Subject(s)
Acute Kidney Injury , Extracellular Vesicles , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Acute Kidney Injury/pathology , Acute Kidney Injury/therapy , Humans , Animals , Male , Reperfusion Injury/pathology , Reperfusion Injury/therapy , Leukocytes, Mononuclear/metabolism , Fibrosis , Apoptosis/drug effects , Neovascularization, Physiologic , MicroRNAs/metabolism , MicroRNAs/genetics , Mice
16.
Theranostics ; 14(9): 3583-3602, 2024.
Article in English | MEDLINE | ID: mdl-38948067

ABSTRACT

Rationale: Mesenchymal stromal cells (MSCs) are considered a promising resource for cell therapy, exhibiting efficacy in ameliorating diverse bone diseases. However, most MSCs undergo apoptosis shortly after transplantation and produce apoptotic extracellular vesicles (ApoEVs). This study aims to clarify the potential role of ApoEVs from apoptotic MSCs in ameliorating osteoporosis and molecular mechanism. Methods: In this study, Dio-labeled bone marrow mesenchymal stem cells (BMSCs) were injected into mice to track BMSCs apoptosis and ApoEVs production. ApoEVs were isolated from BMSCs after inducing apoptosis, the morphology, size distribution, marker proteins expression of ApoEVs were characterized. Protein mass spectrometry analysis revealed functional differences in proteins between ApoEVs and BMSCs. BMSCs were adopted to test the cellular response to ApoEVs. Ovariectomy mice were used to further compare the ability of ApoEVs in promoting bone formation. SiRNA and lentivirus were used for gain and loss-of-function assay. Results: The results showed that BMSCs underwent apoptosis within 2 days after being injected into mice and produce a substantial quantity of ApoEVs. Proteomic analysis revealed that ApoEVs carried a diverse functional array of proteins, and easily traversed the circulation to reach the bone. After being phagocytized by endogenous BMSCs, ApoEVs efficiently promoted the proliferation, migration, and osteogenic differentiation of BMSCs. In an osteoporosis mouse model, treatment of ApoEVs alleviated bone loss and promoted bone formation. Mechanistically, ApoEVs carried Ras protein and activated the Ras/Raf1/Mek/Erk pathway to promote osteogenesis and bone formation in vitro and in vivo. Conclusion: Given that BMSC-derived ApoEVs are high-yield and easily obtained, our data underscore the substantive role of ApoEVs from dying BMSCs to treat bone loss, presenting broad implications for cell-free therapeutic modalities.


Subject(s)
Apoptosis , Extracellular Vesicles , Mesenchymal Stem Cells , Osteogenesis , Osteoporosis , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Mesenchymal Stem Cells/metabolism , Osteoporosis/therapy , Osteoporosis/metabolism , Mice , Female , Osteogenesis/physiology , Cell Differentiation , Mesenchymal Stem Cell Transplantation/methods , Cell Proliferation , Mice, Inbred C57BL , Disease Models, Animal , Ovariectomy , Proteomics , Signal Transduction
17.
J Control Release ; 373: 803-822, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39084466

ABSTRACT

Ischemic stroke-induced mitochondrial dysfunction in the blood-brain barrier-forming brain endothelial cells (BECs) results in long-term neurological dysfunction post-stroke. We previously reported data from a pilot study where intravenous administration of human BEC (hBEC)-derived mitochondria-containing extracellular vesicles (EVs) showed a potential efficacy signal in a mouse middle cerebral artery occlusion (MCAo) model of stroke. We hypothesized that EVs harvested from donor species homologous to the recipient species (e.g., mouse) may improve therapeutic efficacy, and therefore, use of mouse BEC (mBEC)-derived EVs may improve post-stroke outcomes in MCAo mice. We investigated potential differences in the mitochondria transfer of EVs derived from the same species as the recipient cell (mBEC-EVs and recipient mBECs or hBECs-EVs and recipient hBECs) vs. cross-species EVs and recipient cells (mBEC-EVs and recipient hBECs or vice versa). Our results showed that while both hBEC- and mBEC-EVs transferred EV mitochondria, mBEC-EVs outperformed hBEC-EVs in increasing ATP levels and improved recipient mBEC mitochondrial function via increasing oxygen consumption rates. mBEC-EVs significantly reduced brain infarct volume and neurological deficit scores compared to vehicle-injected MCAo mice. The superior therapeutic efficacy of mBEC-EVs in MCAo mice support the continued use of mBEC-EVs to optimize the therapeutic potential of mitochondria-containing EVs in preclinical mouse models.


Subject(s)
Brain , Endothelial Cells , Extracellular Vesicles , Infarction, Middle Cerebral Artery , Ischemic Stroke , Mice, Inbred C57BL , Mitochondria , Animals , Extracellular Vesicles/transplantation , Extracellular Vesicles/metabolism , Mitochondria/metabolism , Humans , Endothelial Cells/metabolism , Ischemic Stroke/therapy , Ischemic Stroke/metabolism , Infarction, Middle Cerebral Artery/therapy , Brain/metabolism , Male , Mice , Cells, Cultured , Blood-Brain Barrier/metabolism
18.
Stem Cell Rev Rep ; 20(7): 1795-1812, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39001965

ABSTRACT

Stem cells-derived extracellular vesicles (SC-EVs) have emerged as promising therapeutic agents for wound repair, recapitulating the biological effects of parent cells while mitigating immunogenic and tumorigenic risks. These EVs orchestrate wound healing processes, notably through modulating angiogenesis-a critical event in tissue revascularization and regeneration. This study provides a comprehensive overview of the multifaceted mechanisms underpinning the pro-angiogenic capacity of EVs from various stem cell sources within the wound microenvironment. By elucidating the molecular intricacies governing their angiogenic prowess, we aim to unravel the mechanistic repertoire underlying their remarkable potential to accelerate wound healing. Additionally, methods to enhance the angiogenic effects of SC-EVs, current limitations, and future perspectives are highlighted, emphasizing the significant potential of this rapidly advancing field in revolutionizing wound healing strategies.


Subject(s)
Extracellular Vesicles , Neovascularization, Physiologic , Stem Cells , Wound Healing , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Stem Cells/metabolism , Stem Cells/cytology , Animals
19.
Stem Cell Rev Rep ; 20(7): 1813-1818, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38976143

ABSTRACT

Regenerative medicine aims to restore, replace, and regenerate human cells, tissues, and organs. Despite significant advancements, many cell therapy trials for cardiovascular diseases face challenges like cell survival and immune compatibility, with benefits largely stemming from paracrine effects. Two promising therapeutic tools have been recently emerged in cardiovascular diseases: extracellular vesicles (EVs) and mitochondrial transfer. Concerning EVs, the first pivotal study with EV-enriched secretome derived from cardiovascular progenitor cells has been done treating heart failure. This first in man demonstrated the safety and feasibility of repeated intravenous infusions and highlighted significant clinical improvements, including enhanced cardiac function and reduced symptoms in heart failure patients. The second study uncovered a novel mechanism of endothelial regeneration through mitochondrial transfer via tunneling nanotubes (TNTs). This research showed that mesenchymal stromal cells (MSCs) transfer mitochondria to endothelial cells, significantly enhancing their bioenergetics and vessel-forming capabilities. This mitochondrial transfer was crucial for endothelial cell engraftment and function, offering a new strategy for vascular regeneration without the need for additional cell types. Combining EV and mitochondrial strategies presents new clinical opportunities. These approaches could revolutionize regenerative medicine, offering new hope for treating cardiovascular and other degenerative diseases. Continued research and clinical trials will be crucial in optimizing these therapies, potentially leading to personalized medicine approaches that enhance patient outcomes.


Subject(s)
Cardiovascular Diseases , Extracellular Vesicles , Mitochondria , Regenerative Medicine , Humans , Regenerative Medicine/methods , Mitochondria/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Cardiovascular Diseases/therapy , Cardiovascular Diseases/pathology , Cardiovascular Diseases/metabolism , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/transplantation , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Animals , Regeneration
20.
Biofabrication ; 16(4)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38986455

ABSTRACT

Over the past three decades, cell therapy development has fallen short of expectations, with many cellular sources demonstrating a 'Janus effect' and raising safety concerns. Extracellular vesicles (EVs), supported by advanced technologies, present a promising avenue in regenerative medicine, offering benefits such as immune tolerance and avoidance of negative aspects associated with cell transplants. Our previous research showcased enhanced and organized subcutaneous vascularization using three-dimensional bioprinted patches containing HUVEC-derived EVs in immunodeficient animal models. In this context, stress conditions on the cells of origin further boosted the EVs' neoangiogenic potential. Since neovascularization is the first regenerative target requiring restoration, the present study aims to complement our previous work by employing an injectable gelatin methacrylate (GelMA) hydrogel functionalized with HUVEC-derived EVs in a pathological condition of acute myocardial infarction. This bioactive hydrogel resulted in reduced fibrosis, improved contractility, and promoted angiogenesis, showing promise in countering tissue deterioration and addressing vascular deficits. Moreover, the molecular characterization of EVs through miRNome and proteomic analyses further supports their potential as bio-additives for hydrogel functionalization. This cell-free approach mitigates immune rejection and oncogenic risks, offering innovative therapeutic advantages.


Subject(s)
Extracellular Vesicles , Human Umbilical Vein Endothelial Cells , Hydrogels , Myocardial Infarction , Neovascularization, Physiologic , Humans , Animals , Myocardial Infarction/therapy , Myocardial Infarction/pathology , Hydrogels/chemistry , Neovascularization, Physiologic/drug effects , Extracellular Vesicles/metabolism , Extracellular Vesicles/chemistry , Extracellular Vesicles/transplantation , Methacrylates/chemistry , Gelatin/chemistry , Injections , Male
SELECTION OF CITATIONS
SEARCH DETAIL