Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Iran J Kidney Dis ; 18(2): 99-107, 2024 03.
Article in English | MEDLINE | ID: mdl-38660698

ABSTRACT

INTRODUCTION: We recently discovered that microvesicles (MVs)  derived from mesenchymal stem cells (MSCs) overexpressing  miRNA-34a can alleviate experimental kidney injury in mice. In  this study, we further explored the effects of miR34a-MV on renal  fibrosis in the unilateral ureteral obstruction (UUO) models.  Methods. Bone marrow MSCs were modified by lentiviruses  overexpressing miR-34a, and MVs were collected from the  supernatants of MSCs. C57BL6/J mice were divided into control,  unilateral ureteral obstruction (UUO), UUO + MV, UUO + miR-34aMV and UUO + miR-34a-inhibitor-MV groups. MVs were injected  to mice after surgery. The mice were then euthanized on day 7  and 14 of modeling, and renal tissues were collected for further  analyses by Hematoxylin and eosin, Masson's trichrome,  and Immunohistochemical (IHC) staining.  Results. The UUO + MV group exhibited a significantly reduced  degree of renal interstitial fibrosis with inflammatory cell infiltration,  tubular epithelial cell atrophy, and vacuole degeneration compared  with the UUO group. Surprisingly, overexpressing miR-34a enhanced  these effects of MSC-MV on the UUO mice.  Conclusion. Our study demonstrates that miR34a further enhances  the effects of MSC-MV on renal fibrosis in mice through the  regulation of epithelial-to-mesenchymal transition (EMT) and  Notch pathway. miR-34a may be a candidate molecular therapeutic  target for the treatment of renal fibrosis. DOI: 10.52547/ijkd.7673.


Subject(s)
Cell-Derived Microparticles , Kidney Diseases , Kidney , Mesenchymal Stem Cells , MicroRNAs , Animals , Male , Mice , Cell-Derived Microparticles/genetics , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/transplantation , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Kidney/pathology , Kidney/metabolism , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics , Signal Transduction , Ureteral Obstruction
2.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34210091

ABSTRACT

(1) Background: We established a new bladder ischemia rat model through bilateral partial iliac arterial occlusion (BPAO) and investigated the therapeutic effect of adipose-derived stem cells (ADSCs) and ADSC-derived microvesicles (MVs); (2) Methods: The study included four groups: (1) sham, (2) BPAO, (3) BPAO + ADSCs, and (4) BPAO + ADSC-derived MVs. Female Wistar rats with BPAO were injected with ADSCs or ADSC-derived MVs through the femoral artery. Doppler flowmetry and real-time laser speckle contrast imaging were performed to quantify blood flow in the common iliac arteries and bladder microcirculation. A 24-h behavior study and transcystometrogram were conducted after 2 weeks. Bladder histology, immunostaining, and lipid peroxidation assay were performed. The expressions of P2X2, P2X3, M2, and M3 receptors and nerve growth factor (NGF) were evaluated; (3) Results: BPAO significantly reduced bladder microcirculation, intercontraction interval (ICI), and bladder volume and increased the amplitude of nonvoiding contraction, neutrophil infiltration, and malondialdehyde and NGF levels. ADSCs and ADSC-derived MVs significantly ameliorated these effects. The results of Western blot showed that the BPAO group exhibited the highest expression of M3 and P2X2 receptors. ADSCs significantly attenuated the expressions of M2 and P2X2 receptors. ADSC-derived MVs significantly attenuated the expressions of M3 and P2X2 receptors; (4) Conclusions: ADSCs and ADSC-derived MVs ameliorated the adverse effects of BPAO including bladder overactivity, bladder ischemia, and oxidative stress. Inflammation, muscarinic signaling, purinergic signaling, and NGF might be involved in the therapeutic mechanism.


Subject(s)
Adipose Tissue/cytology , Adult Stem Cells/transplantation , Cell-Derived Microparticles/transplantation , Urinary Bladder, Overactive/therapy , Adult Stem Cells/cytology , Animals , Arterial Occlusive Diseases/complications , Arterial Occlusive Diseases/therapy , Cell-Derived Microparticles/physiology , Disease Models, Animal , Female , Iliac Artery/pathology , Ischemia/etiology , Ischemia/therapy , Rats , Rats, Wistar , Urinary Bladder/pathology , Urinary Bladder, Overactive/etiology
3.
Nat Nanotechnol ; 16(8): 942-951, 2021 08.
Article in English | MEDLINE | ID: mdl-34140674

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has grown into a global pandemic, and only a few antiviral treatments have been approved to date. Angiotensin-converting enzyme 2 (ACE2) plays a fundamental role in SARS-CoV-2 pathogenesis because it allows viral entry into host cells. Here we show that ACE2 nanodecoys derived from human lung spheroid cells (LSCs) can bind and neutralize SARS-CoV-2 and protect the host lung cells from infection. In mice, these LSC-nanodecoys were delivered via inhalation therapy and resided in the lungs for over 72 h post-delivery. Furthermore, inhalation of the LSC-nanodecoys accelerated clearance of SARS-CoV-2 mimics from the lungs, with no observed toxicity. In cynomolgus macaques challenged with live SARS-CoV-2, four doses of these nanodecoys delivered by inhalation promoted viral clearance and reduced lung injury. Our results suggest that LSC-nanodecoys can serve as a potential therapeutic agent for treating COVID-19.


Subject(s)
COVID-19 Drug Treatment , Lung Injury/prevention & control , Nanostructures/administration & dosage , SARS-CoV-2/drug effects , Administration, Inhalation , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/virology , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/transplantation , Disease Models, Animal , Humans , Lung Injury/virology , Macaca fascicularis , Mice , Protein Binding , SARS-CoV-2/metabolism , Spheroids, Cellular/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Viral Load/drug effects
4.
Int J Biol Sci ; 17(5): 1339-1352, 2021.
Article in English | MEDLINE | ID: mdl-33867850

ABSTRACT

Treatment for spinal cord injury (SCI) remains a challenge worldwide, and inflammation is a major cause of secondary injury after SCI. Peripheral macrophages (PMs) have been verified as a key factor that exert anti-inflammatory effects after SCI, but the mechanism is unidentified. As local macrophages, microglia also exert significant effects after SCI, especially polarization. Exosomes show source cell-like biological functions to target cells and have been the subject of much research in recent years. Thus, we hypothesized the PM-derived exosomes (PM-Exos) play an important role in signal transmission with local microglia and can be used therapeutic agents for SCI in a series of in vivo and in vitro studies. For the in vivo experiment, three groups of Sprague-Dawley (SD) rats subjected to spinal cord contusion injury were injected with 200 µg/ml PM-Exos, 20 µg/ml PM-Exos or PBS via the tail vein. Recovery of the rats and of spinal cord function were observed. In vitro, we investigated the potential anti-inflammatory mechanism of PM-Exos and evaluated microglial autophagy, anti-inflammatory type microglia polarization and the upstream signaling pathway. The results showed that spinal cord function and recovery were better in the PM-Exo groups than the control group. In the in vitro study, microglial autophagy levels and the expression of anti-inflammatory type microglia were higher in the experimental groups than the control group. Moreover, the expression of proteins related to the PI3K/AKT/mTOR autophagic signaling pathway was suppressed in the PM-Exo groups. PM-Exos have a beneficial effect in SCI, and activation of microglial autophagy via inhibition of the PI3K/AKT/mTOR signaling pathway, enhancing the polarization of anti-inflammatory type microglia, that may play a major role in the anti-inflammatory process.


Subject(s)
Autophagy/immunology , Exosomes , Inflammation , Macrophages/immunology , Microglia , Spinal Cord Injuries , Animals , Cell Polarity/physiology , Cell-Derived Microparticles/immunology , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/transplantation , Exosomes/immunology , Exosomes/metabolism , Exosomes/transplantation , Inflammation/metabolism , Inflammation/therapy , Microglia/immunology , Microglia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Signal Transduction , Spinal Cord Injuries/immunology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome
5.
Hum Cell ; 34(4): 1103-1113, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33860459

ABSTRACT

The intraoperative ischemia in partial nephrectomy (PN) often leads to postoperative renal function impairment and fibrosis, which can be regulated by macrophage polarization. We have previously demonstrated that microvesicles derived from human Wharton's Jelly mesenchymal stromal cells (hWJMSC-MVs) attenuated renal ischemia-induced renal fibrosis and contained a substantial quantity of hepatocyte growth factor (HGF). Herein, we investigated whether MSC-MVs regulate macrophage polarization and ameliorate renal fibrosis following ischemia-PN via transferring HGF. A rat model of ischemia-PN was established by 45 min of left renal ischemia followed by removal of 1/3 upper left kidney. MSC-MVs were injected through the tail vein immediately after ischemia. Renal injury biomarkers were measured and histologic analysis was performed to analyze renal injury. A co-culture model of THP-1 macrophages and MSC-MVs was utilized. The expression of M1 markers and M2 markers were determined to evaluate macrophage polarization. MSC-MV administration significantly ameliorated renal inflammation, lesions, and fibrosis in ischemia-PN rats, and promoted M2 macrophage polarization both in rat remnant renal tissues and LPS-treated THP-1 cells. These effects of MSC-MVs were compromised when HGF expression was downregulated in MSC-MVs. Collectively, MSC-MVs promote M2 macrophage polarization and attenuate renal fibrosis following ischemia-PN via transferring HGF.


Subject(s)
Cell-Derived Microparticles/physiology , Gene Expression , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Kidney/pathology , Macrophages/physiology , Mesoderm/cytology , Nephrectomy/adverse effects , Nephrectomy/methods , Stromal Cells/physiology , Umbilical Cord/cytology , Wharton Jelly/cytology , Animals , Cell Polarity , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/transplantation , Disease Models, Animal , Fibrosis , Humans , Intraoperative Complications/etiology , Ischemia/etiology , Kidney/blood supply , Kidney/metabolism , Male , Postoperative Complications/etiology , Rats, Sprague-Dawley , Stromal Cells/metabolism , Stromal Cells/transplantation , THP-1 Cells
6.
Front Immunol ; 12: 651109, 2021.
Article in English | MEDLINE | ID: mdl-33790914

ABSTRACT

Mesenchymal stromal cells (MSC) are a promising therapy for inflammatory diseases. However, MSC are large and become trapped in the lungs after intravenous infusion, where they have a short survival time. To steer MSC immunoregulatory therapy beyond the lungs, we generated nm-sized particles from MSC membranes (membrane particles, MP), which have immunomodulatory properties, and investigated their internalization and mode of interaction in macrophages subtypes and human umbilical vein endothelial cells (HUVEC) under control and inflammatory conditions. We found that macrophages and HUVEC take up MP in a dose, time, and temperature-dependent manner. Specific inhibitors for endocytotic pathways revealed that MP internalization depends on heparan sulfate proteoglycan-, dynamin-, and clathrin-mediated endocytosis but does not involve caveolin-mediated endocytosis. MP uptake also involved the actin cytoskeleton and phosphoinositide 3-kinase, which are implicated in macropinocytosis and phagocytosis. Anti-inflammatory M2 macrophages take up more MP than pro-inflammatory M1 macrophages. In contrast, inflammatory conditions did not affect the MP uptake by HUVEC. Moreover, MP induced both anti- and pro-inflammatory responses in macrophages and HUVEC by affecting gene expression and cell surface proteins. Our findings on the mechanisms of uptake of MP under different conditions help the development of target-cell specific MP therapy to modulate immune responses.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cell-Derived Microparticles/immunology , Mesenchymal Stem Cells/cytology , Cell-Derived Microparticles/transplantation , Cells, Cultured , Dose-Response Relationship, Immunologic , Healthy Volunteers , Human Umbilical Vein Endothelial Cells , Humans , Macrophages/immunology , Mesenchymal Stem Cells/immunology , Phagocytosis/immunology , Pinocytosis/immunology , Primary Cell Culture , Subcutaneous Fat/cytology
7.
PLoS One ; 16(3): e0248415, 2021.
Article in English | MEDLINE | ID: mdl-33730089

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease with limited treatment options in which the telomere shortening is a strong predictive factor of poor prognosis. Mesenchymal stromal cells (MSC) administration is probed in several experimental induced lung pathologies; however, MSC might stimulate fibrotic processes. A therapy that avoids MSC side effects of transformation would be an alternative to the use of living cells. Membranes particles (MP) are nanovesicles artificially generated from the membranes of MSC containing active enzymes involved in ECM regeneration. We aimed to investigate the anti-fibrotic role of MP derived from MSC in an in vitro model of pulmonary fibrosis. METHODS: Epithelial cells (A549) and lung fibroblasts, from IPF patients with different telomere length, were co-cultured with MP and TGF-ß for 48h and gene expression of major pro-fibrotic markers were analyzed. RESULTS: About 90% of both types of cells effectively took up MP without cytotoxic effects. MP decreased the expression of profibrotic proteins such as Col1A1, Fibronectin and PAI-1, in A549 cells. In fibroblasts culture, there was a different response in the inhibitory effect of MP on some pro-fibrotic markers when comparing fibroblast from normal telomere length patients (FN) versus short telomere length (FS), but both types showed an inhibition of Col1A1, Tenascin-c, PAI-1 and MMP-1 gene expression after MP treatment. CONCLUSIONS: MP conserve some of the properties attributed to the living MSC. This study shows that MP target lung cells, via which they may have a broad anti-fibrotic effect.


Subject(s)
Cell-Derived Microparticles/transplantation , Idiopathic Pulmonary Fibrosis/therapy , Mesenchymal Stem Cells/cytology , Nanoparticles/therapeutic use , Primary Cell Culture/methods , A549 Cells , Adult , Aged , Coculture Techniques , Female , Fibroblasts , Gene Expression Regulation , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/cytology , Lung/pathology , Male , Mesenchymal Stem Cell Transplantation/adverse effects , Middle Aged , Subcutaneous Fat/cytology , Telomere Shortening
8.
Genes (Basel) ; 12(2)2021 01 27.
Article in English | MEDLINE | ID: mdl-33513776

ABSTRACT

Exosomes are packaged with a variety of cellular cargo including RNA, DNA, lipids and proteins. For several decades now there has been ongoing debate as to what extent exosomes are the garbage bin of the cell or if these entities function as a distributer of cellular cargo which acts in a meaningful mechanistic way on target cells. Are the contents of exosomes unwanted excess cellular produce or are they selective nucleic acid packaged nanoparticles used to communicate in a paracrine fashion? Overexpressed RNAs and fragments of DNA have been shown to collect into exosomes which are jettisoned from cells in response to particular stimuli to maintain homeostasis suggesting exosomes are functional trash bins of the cell. Other studies however have deciphered selective packaging of particular nucleic acids into exosomes. Nucleic acids packaged into exosomes are increasingly reported to exert transcriptional control on recipient cells, supporting the notion that exosomes may provide a role in signaling and intracellular communication. We survey the literature and conclude that exosomes are multifunctional entities, with a plethora of roles that can each be taken advantage to functionally modulate cells. We also note that the potential utility of developing exosomes as a next generation genetic therapy may in future transform cellular therapies. We also depict three models of methodologies which can be adopted by researchers intending to package nucleic acid in exosomes for developing gene and cell therapy.


Subject(s)
Cell- and Tissue-Based Therapy , Exosomes/metabolism , Genetic Therapy , Animals , Bioengineering/methods , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/transplantation , DNA/administration & dosage , DNA/genetics , Drug Carriers , Exosomes/transplantation , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Therapy/trends , Humans , Nanoparticles , RNA/administration & dosage , RNA/genetics
9.
Theranostics ; 11(5): 2410-2423, 2021.
Article in English | MEDLINE | ID: mdl-33500733

ABSTRACT

Rationale: Abnormal migration of vascular smooth muscle cells (VSMCs) from the media to the interior is a critical process during the intimal restenosis caused by vascular injury. Here, we determined the role of platelet-derived microvesicles (PMVs) released by activated platelets in VSMC migration. Methods: A percutaneous transluminal angioplasty balloon dilatation catheter was used to establish vascular intimal injury. Collagen I was used to activate PMVs, mimicking collagen exposure during intimal injury. To determine the effects of PMVs on VSMC migration in vitro, scratch wound healing assays were performed. Fluorescence resonance energy transfer was used to detect variations of calcium dynamics in VSMCs. Results: Morphological results showed that neointimal hyperplasia was markedly increased after balloon injury of the carotid artery in rats, and the main component was VSMCs. PMVs significantly promoted single cell migration and wound closure in vitro. Fluorescence resonance energy transfer revealed that PMVs induced temporal and dynamic calcium oscillations in the cytoplasms of VSMCs. The influx of extracellular calcium, but not calcium from intracellular stores, was involved in the process described above. The channel antagonist GSK219 and specific siRNA revealed that a membrane calcium channel, transient receptor potential vanilloid 4 (TRPV4), participated in the calcium oscillations and VSMC migration induced by PMVs. Conclusions: TRPV4 participated in the calcium oscillations and VSMC migration induced by PMVs. PMVs and the related molecules might be novel therapeutic targets for vascular remodeling during vascular injury.


Subject(s)
Blood Platelets/metabolism , Calcium Signaling , Cell Movement , Cell-Derived Microparticles/transplantation , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Animals , Cell Proliferation , Cells, Cultured , Male , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Rats , Rats, Sprague-Dawley , TRPV Cation Channels/genetics
10.
Arterioscler Thromb Vasc Biol ; 41(1): 87-96, 2021 01.
Article in English | MEDLINE | ID: mdl-33028092

ABSTRACT

Extracellular vesicles (EVs) are a means of cell-to-cell communication and can facilitate the exchange of a broad array of molecules between adjacent or distant cells. Platelets are anucleate cells derived from megakaryocytes and are primarily known for their role in maintaining hemostasis and vascular integrity. Upon activation by a variety of agonists, platelets readily generate EVs, which were initially identified as procoagulant particles. However, as both platelets and their EVs are abundant in blood, the role of platelet EVs in hemostasis may be redundant. Moreover, findings have challenged the significance of platelet-derived EVs in coagulation. Looking beyond hemostasis, platelet EV cargo is incredibly diverse and can include lipids, proteins, nucleic acids, and organelles involved in numerous other biological processes. Furthermore, while platelets cannot cross tissue barriers, their EVs can enter lymph, bone marrow, and synovial fluid. This allows for the transfer of platelet-derived content to cellular recipients and organs inaccessible to platelets. This review highlights the importance of platelet-derived EVs in physiological and pathological conditions beyond hemostasis.


Subject(s)
Blood Platelets/metabolism , Cell Communication , Cell-Derived Microparticles/metabolism , Hemostasis , Platelet Activation , Animals , Bone Marrow/metabolism , Cell-Derived Microparticles/transplantation , Humans , Inflammation Mediators/blood , Lymph/metabolism , Synovial Fluid/metabolism
11.
J Cardiovasc Pharmacol ; 76(6): 650-657, 2020 12.
Article in English | MEDLINE | ID: mdl-33105323

ABSTRACT

The role of stem cells in augmenting reparative processes in the heart after ischemic injury has been successfully demonstrated in small and large animal models. However, the outcomes of cell therapy in clinical trials have been somewhat variable, with overall effects of autologous stem cell therapies demonstrating a modest improvement in cardiac structure and function. How stem cells repair the heart after cardiac injury is still not well understood. Most recent studies suggest that adult derived stem cells act primarily through paracrine signaling to exert beneficial effects, including modulation of immune response, stimulation of new blood vessel formation, or by inducing mature myocytes to transiently reenter the cell cycle, rather than robust direct differentiation of the transplanted cells into myocytes. In addition, data from multiple laboratory results confirmed clearance of stem cells themselves within a few days still leading to functional benefits further confirming the role of paracrine signaling in augmenting cardiac reparative processes rather than direct differentiation of cells. These findings rapidly evolved the field of extracellular vesicles specifically microvesicles (MVs) as they are active hubs of autocrine, paracrine, and endocrine signaling targeting different biological processes. The beneficial effects seen after stem cell transplantation could be linked to the cardioprotective factors packaged in the MVs secreted from stem cells. Therefore, stem cell MVs provide a new avenue for the treatment of cardiovascular disease through a multitude of mechanisms including cellular communication within the stem cell niches, delivery of genetic information, regulation of the immune system in the heart, and stimulation of angiogenesis which will be discussed in this review.


Subject(s)
Cardiovascular Diseases/surgery , Cell-Derived Microparticles/transplantation , Myocardium/pathology , Regeneration , Stem Cell Transplantation , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cell-Derived Microparticles/metabolism , Humans , Myocardium/metabolism , Neovascularization, Physiologic , Recovery of Function , Signal Transduction , Stem Cell Niche , Treatment Outcome
12.
Stem Cells Dev ; 29(12): 747-754, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32380908

ABSTRACT

This prospective nonrandomized open-label cohort study addresses the safety and efficacy of exosomes (ExoFlo™) derived from allogeneic bone marrow mesenchymal stem cells as treatment for severe COVID-19. During April 2020, ExoFlo was provided to 24 SARS-CoV-2 polymerase chain reaction-positive patients at a single hospital center, all of whom met criteria for severe COVID-19 as well as moderate-to-severe acute respiratory distress syndrome. Patients received a single 15 mL intravenous dose of ExoFlo and were evaluated for both safety and efficacy from days 1 to 14 post-treatment. All safety endpoints were met with no adverse events observed within 72 h of ExoFlo administration. A survival rate of 83% was observed. In total, 17 of 24 (71%) patients recovered, 3 of 24 (13%) patients remained critically ill though stable, and 4 of 24 (16%) patients expired for reasons unrelated to the treatment. Overall, after one treatment, patients' clinical status and oxygenation improved with an average pressure of arterial oxygen to fraction of inspired oxygen ratio (PaO2/FiO2) increase of 192% (P < 0.001). Laboratory values revealed significant improvements in absolute neutrophil count [mean reduction 32% (P value <0.001)] and lymphopenia with average CD3+, CD4+, and CD8+ lymphocyte counts increasing by 46% (P < 0.05), 45% (P < 0.05), and 46% (P < 0.001), respectively. Likewise, acute phase reactants declined, with mean C-reactive protein, ferritin, and D-dimer reduction of 77% (P < 0.001), 43% (P < 0.001), and 42% (P < 0.05), respectively. In conclusion, owing to its safety profile, capacity to restore oxygenation, downregulate cytokine storm, and reconstitute immunity, ExoFlo is a promising therapeutic candidate for severe COVID-19. Future randomized controlled trials (RCTs) are needed to determine ExoFlo therapeutic potential.


Subject(s)
Bone Marrow Cells/cytology , Coronavirus Infections/therapy , Critical Illness/therapy , Exosomes/transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Pneumonia, Viral/therapy , Adult , Aged , Aged, 80 and over , Betacoronavirus/pathogenicity , COVID-19 , Cell-Derived Microparticles/transplantation , Cohort Studies , Coronavirus Infections/mortality , Critical Illness/mortality , Female , Humans , Male , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/ultrastructure , Middle Aged , Pandemics , Pneumonia, Viral/mortality , SARS-CoV-2 , Severity of Illness Index , Treatment Outcome
13.
FEBS J ; 287(23): 5196-5217, 2020 12.
Article in English | MEDLINE | ID: mdl-32167224

ABSTRACT

Intimal injury is an early stage of several cardiovascular diseases. Endothelial progenitor cells (EPCs) play a significant role in endothelial repair following vascular injury. Once the intima is damaged, EPCs are mobilized from the bone marrow to the injury site. Meanwhile, the injury to the intimal surface triggers platelet degranulation, aggregation, and adhesion to the damaged endothelium, and exposed collagen stimulates platelet to secrete platelet-derived microvesicles (PMVs). However, the role of PMVs in EPC function during this process remains unknown. In an in vivo study, EPCs and platelets were found to adhere to the injury site in Sprague-Dawley (SD) rat vascular injury model. In vitro, collagen stimulation induced the release of PMVs, and collagen-activated PMVs (ac.PMVs) significantly promoted EPC proliferation. Transforming growth factor-ß1 (TGF-ß1) content was increased in ac.PMVs. Activated PMVs significantly upregulated Smad3 phosphorylation in EPCs and increased Smad3 nuclear translocation from the cytoplasm. TGF-ß1 knockdown ac.PMVs downregulated EPC proliferation. Recombinant TGF-ß1 enhanced EPC proliferation. The TGF-ß1 inhibitor SB431542 significantly repressed the intracellular signal triggered by ac.PMVs. Furthermore, the Smad3-specific phosphorylation inhibitor SIS3 effectively reversed the cell proliferation induced by ac.PMVs. Smad3 translocated to the nucleus and enhanced EPC proliferation via its downstream genes tenascin C (TNC), CDKN1A, and CDKN2A. r-TGF-ß1 promoted reendothelialization and EPC proliferation in vivo. Our data demonstrate that activated PMVs deliver TGF-ß1 from collagen-activated platelets to EPCs, which in turn activates Smad3 phosphorylation and regulates TNC, CDKN1A, and CDKN2A expression to promote EPC proliferation, suggesting that PMVs act as a key transporter and a potential therapeutic target for vascular injury.


Subject(s)
Blood Platelets/metabolism , Carotid Artery Injuries/therapy , Cell Proliferation , Cell-Derived Microparticles/transplantation , Endothelial Progenitor Cells/cytology , Transforming Growth Factor beta1/metabolism , Tunica Intima/metabolism , Animals , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Carotid Intima-Media Thickness , Cell Differentiation , Cell-Derived Microparticles/metabolism , Cells, Cultured , Male , Rats , Rats, Sprague-Dawley , Signal Transduction , Tunica Intima/injuries
14.
Cells ; 9(2)2020 02 12.
Article in English | MEDLINE | ID: mdl-32059493

ABSTRACT

Atherosclerosis and cardiovascular disease development is the outcome of intermediate processes where endothelial dysfunction and vascular inflammation are main protagonists. Cell-derived microvesicles (MVs), endothelial progenitor cells (EPCs), and circulating microRNAs (miRNAs) are known as biomarkers and potential regulators for atherosclerotic vascular disease, but their role in the complexity of the inflammatory process and in the mechanism of vascular restoration is far from clear. We aimed to evaluate the biological activity and functional role of MVs, in particular of the EPCs-derived MVs (MVEs), of healthy origins in reducing atherosclerotic vascular disease development. The experiments were performed on hamsters divided into the following groups: simultaneously hypertensive-hyperlipidemic (HH group) by combining two feeding conditions for 4 months; HH with retro-orbital sinus injection containing 1 × 105 MVs or MVEs from control hamsters, one dose per month for 4 months of HH diet, to prevent atherosclerosis (HH-MVs or HH-MVEs group); and controls (C group), age-matched normal healthy animals. We found that circulating MV and MVE transplantation of healthy origins significantly reduces atherosclerosis development via (1) the mitigation of dyslipidemia, hypertension, and circulating EPC/cytokine/chemokine levels and (2) the structural and functional remodeling of arterial and left ventricular walls. We also demonstrated that (1) circulating MVs contain miRNAs; this was demonstrated by validating MVs and MVEs as transporters of Ago2-miRNA, Stau1-miRNA, and Stau2-miRNA complexes and (2) MV and MVE administration significantly protect against atherosclerotic cardiovascular disease via transfer of miR-223, miR-21, miR-126, and miR-146a to circulating late EPCs. It should be mentioned that the favorable effects of MVEs were greater than those of MVs. Our findings suggest that allogenic MV and MVE administration of healthy origins could counteract HH diet-induced detrimental effects by biologically active miR-10a, miR-21, miR-126, and miR-146a transfer to circulating EPCs, mediating their vascular repair function in atherosclerosis processes.


Subject(s)
Atherosclerosis/prevention & control , Cell-Derived Microparticles/transplantation , Endothelial Progenitor Cells/metabolism , Administration, Intravenous , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Atherosclerosis/pathology , Blood Pressure , Cell-Derived Microparticles/metabolism , Chemokines/blood , Chemokines/metabolism , Cricetinae , Cytokines/blood , Cytokines/metabolism , Diet, High-Fat , Endothelial Progenitor Cells/cytology , Endothelium/anatomy & histology , Endothelium/ultrastructure , Heart Rate , Male , MicroRNAs/metabolism , Transplantation, Homologous , Triglycerides/blood , Ventricular Remodeling
15.
J Cell Physiol ; 235(2): 706-717, 2020 02.
Article in English | MEDLINE | ID: mdl-31254289

ABSTRACT

Mesenchymal stem cells (MSCs) have been revealed to hold great potential for the development of new treatment approaches for various diseases. However, the clinical use of these cells is limited due to their tumorigenic effects. The therapeutic benefits of MSCs are largely dependent on paracrine factors including extracellular vesicles (EVs). EVs are nano-sized bilayer membrane structures containing lipids, microRNAs and proteins which play key roles in cell-to-cell communications. Because of their lower immunogenicity, tumorigenicity, and easier management, EVs have emerged as a new promising alternative to whole-cell therapy. Therefore, this paper reviews current preclinical studies on the use of EVs derived from human umbilical cord MSCs (hucMSCs) as a therapeutic approach in treatment of several diseases including neurological, cardiovascular, liver, kidney, and bone diseases as well as the cutaneous wound, inflammatory bowel disease, cancers, infertility, and other disorders.


Subject(s)
Exosomes/metabolism , Exosomes/transplantation , Fetal Blood/cytology , Mesenchymal Stem Cells/metabolism , Animals , Cell Line, Tumor , Cell- and Tissue-Based Therapy/adverse effects , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/transplantation , Disease Models, Animal , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Mice , Rats , Umbilical Cord/cytology
16.
Stem Cell Res Ther ; 10(1): 394, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31852535

ABSTRACT

BACKGROUND: In athlete horses, suspensory ligament (SL) injuries are the most common cause of lameness. Healing of SL injury is still problematic, and even proper rehabilitation and pharmacological therapy do not guarantee returning to the initial performance level. In our previous studies, we have shown that a combination of 5-azacytidine (AZA) and resveratrol (RES) exerts beneficial, rejuvenating effects on metabolic syndrome derived adipose-derived stem cells (ASCs). Thus, in the presented research, we investigate whether not only rejuvenated ASC but also microvesicles (MVsAZA/RES) secreted by them possess enhanced regenerative properties in SL injury. METHODS: In the presented study, a 6-year-old Dutch Warmblood gelding, working in jumping, was diagnosed with SL injury using ultrasonography, Doppler, real-time elastography and thermography. As a therapeutic strategy, the affected animal was treated with extracellular microvesicles derived from ASC treated with the combination of 5-azacytydine (AZA) and resveratrol (RES) (MVsAZA/RES). RESULTS: First, anti-apoptotic effects of MVsAZA/RES were tested in co-culture with metabolic syndrome derived ASC. The proliferation of cells and expression of pro-apoptotic genes were investigated. Then, MVsAZA/RES were injected directly into the injured SL of the Dutch Warmblood gelding. In vitro assays revealed that MVsAZA/RES enhance the proliferation of ASC and exert an anti-apoptotic effect. In the affected horse, the application of MVsAZA/RES resulted in increased lesion filling and improvement of angiogenesis and elasticity in injured tissue. CONCLUSIONS: As MVsAZA/RES mimic several of the biological actions exerted by ASC, they have become an alternative for stem cell-based therapies and can be effectively applied for the treatment of SL injury in horses.


Subject(s)
Azacitidine/pharmacology , Cell-Derived Microparticles/transplantation , Horse Diseases/therapy , Ligaments/injuries , Mesenchymal Stem Cells/drug effects , Resveratrol/pharmacology , Animals , Cell-Derived Microparticles/metabolism , Elasticity Imaging Techniques , Horse Diseases/pathology , Horses , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Ultrasonography , Wound Healing
17.
J Nutr Biochem ; 74: 108242, 2019 12.
Article in English | MEDLINE | ID: mdl-31665654

ABSTRACT

Breast cancer is one of the most frequent and malignant types of cancer in women, with an increasing morbidity and mortality rate; in particular, treatment of triple negative breast cancer remains a challenge, since the efforts made with targeted therapies were ineffective. Among surrounding cells influencing the biology of cancer cells, platelets are recognizing as novel players. Activated platelets release microvesicles (MVs) that, once delivered to cancer cells, modulate signaling pathways related to cell growth and dissemination; among factors contained in platelet-derived MVs, microRNAs are highly involved in cancer development. The growing interest in ω3 and ω6 polyunsaturated fatty acids (PUFAs) as adjuvants in anti-cancer therapy prompted us to investigate the ability of arachidonic acid (AA) and docosahexaenoic acid (DHA) to modulate MV biological functions. AA induced differential enhancement of platelet-specific microRNAs (miR-223 and miR-126), an effect further enhanced by the presence of DHA. MVs can be delivered to and microRNAs internalized by breast cancer cells, although with different efficiency; analysis of kinetics of MV delivery, indeed, suggested that tumor cells fine-tune the uptake of specific microRNA. Finally, we demonstrated that physiological delivery of platelet miR-223 and miR-126 induced cellular effects in breast cancer cells, including cell cycle arrest, inhibition of migration and sensitivity to cisplatin. These results have been confirmed by exogenous expression of miR-223 and miR-126 through transient transfection experiments. Our preliminary data suggest that ω6/ω3-PUFA supplementation, by modulating microRNA delivery, enhances platelet anti-tumor activities, thus opening new avenues for add-on therapies in cancer patients.


Subject(s)
Arachidonic Acid/pharmacology , Blood Platelets/drug effects , Breast Neoplasms/genetics , Cell-Derived Microparticles/genetics , Docosahexaenoic Acids/pharmacology , Antineoplastic Agents/pharmacology , Blood Platelets/cytology , Blood Platelets/physiology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell-Derived Microparticles/transplantation , Cisplatin/pharmacology , Dietary Supplements , Female , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Humans , MicroRNAs/genetics
18.
Int J Biochem Cell Biol ; 114: 105570, 2019 09.
Article in English | MEDLINE | ID: mdl-31302227

ABSTRACT

Skin wound healing is a complex physiological process that maintains the integrity of the skin tissues, involving a variety of distinct cell types and signaling molecules. The specific signaling pathways or extracellular cues that govern the healing processes remain elusive. Microvesicles (MVs) have recently emerged as critical mediators of cell communication by delivery of genetic materials to target cells. In this study, we found the direct delivery of HEKa-MVs expressing miR-21 mimics significantly promoted the healing of skin wound in diabetic rats. In-depth studies showed that MV miR-21 promoted fibroblast migration, differentiation, and contraction, induced a pro-angiogenic process of endothelial cells and mediated a pro-inflammatory response. Mechanically, MV miR-21 might target specific essential effector mRNA in fibroblasts such as MMP-1, MMP-3, TIMP3, and TIMP4 to increase MMPs expression and enzymatic activities. Moreover, MV miR-21 regulated ɑ-SMA and N-cadherin to induce fibroblast-myofibroblast differentiation. MV miR-21 up-regulated the IL-6 and IL-8 expressions and their secretion to amplify the immune response. Furthermore, MV miR-21 down-regulated PTEN and RECK in protein level, and activate MAPK/ERK signaling cascade, thereby promoting fibroblast functions. Thus, our study has provided for the first time the basis for the potential application of HEKa-MVs, and MV miR-21 in particular for wound healing.


Subject(s)
Cell-Derived Microparticles/transplantation , Diabetes Mellitus, Experimental/therapy , Fibroblasts/metabolism , Keratinocytes/metabolism , MicroRNAs/pharmacology , Neovascularization, Physiologic , Skin/injuries , Wound Healing , Wounds and Injuries/therapy , Animals , Cell Line , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Fibroblasts/pathology , Humans , Keratinocytes/pathology , Male , Rats , Rats, Sprague-Dawley , Skin/blood supply , Skin/metabolism , Skin/pathology , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
19.
Int J Biochem Cell Biol ; 112: 114-122, 2019 07.
Article in English | MEDLINE | ID: mdl-31100425

ABSTRACT

Several studies have highlighted the underlying role of mesenchymal stem cells microvesicles (MSC-MVs) in acute lung injury (ALI). Hepatocyte growth factor (HGF) derived from MSC-MVs is partly involved in their therapeutic effects; however, the detailed mechanism remains unclear. MVs were isolated from human Wharton's Jelly MSCs. The rat model of ALI was established by intratracheal instillation of bleomycin (BLM). A co-culture model of alveolar epithelial cells or pulmonary endothelial cells and MSC-MVs was utilized. Total protein content in bronchoalveolar lavage fluid (BALF) was determined by bicinchoninic acid method. White blood cell (WBC) and neutrophil in BALF were counted. ELISA was used for the determination of cytokines and HGF in BALF. Apoptosis was determined by TUNEL assay and Annexin V-FITC/PI staining as well as caspase-3 activity detection. HE and Masson staining of lung tissues was used for histopathology analysis. The expression of HGF and proteins involved in the PI3K/AKT/mTOR pathway were measured by quantitative Real-Time PCR (qRT-PCR) and western blotting. Treatment with MSC-MVs significantly inhibited BLM-induced apoptosis and fibrosis in lung tissues and PI3K/AKT/mTOR activation, which was reversed by HGF mRNA deficient MVs. Intriguingly, these effects were completely abrogated by PI3K inhibitor. The therapeutic effect of MSC-MVs in ALI was partly mediated through HGF mRNA.


Subject(s)
Acute Lung Injury/therapy , Cell-Derived Microparticles/transplantation , Hepatocyte Growth Factor/metabolism , Mesenchymal Stem Cells/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Bleomycin/adverse effects , Bleomycin/pharmacology , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , Humans , Male , Mesenchymal Stem Cells/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
20.
J Cardiovasc Pharmacol Ther ; 24(5): 474-483, 2019 09.
Article in English | MEDLINE | ID: mdl-31035782

ABSTRACT

BACKGROUND: Uncontrollable bleeding is a major cause of mortality and morbidity worldwide. Effective hemostatic agents are urgently needed. Red cell microparticles (RMPs) are a highly promising hemostatic agent. This study evaluated the safety profile of RMPs preliminary to clinical trial. METHODS AND RESULTS: RMPs were prepared from type O+ human red blood cell by high-pressure extrusion. Male rats were treated with RMPs either a 1 × bolus, or 4 × or 20 × administered over 60 minutes. The vehicle-treated group was used as a control. Effects on physiological parameters were evaluated; namely, blood pressure, body and head temperature, hematocrit, and blood gases. We did not observe any adverse effects of RMPs on these physiological parameters. In addition, brain, heart, and lungs of rats treated with 4 × dose (bolus followed by infusion over 60 minutes) or vehicle were examined histologically for signs of thrombosis or other indications of toxicity. No thrombosis or indications of toxicity in brain, heart, or lungs were observed. Studies revealed that RMPs were distributed mainly in liver, spleen, and lymph nodes, and were potentially excreted through the kidneys. CONCLUSIONS: Our study indicates that RMP administration appears not to have any negative impact on the parameters studied and did not produce thrombosis in heart, brain, and lungs. However, more detailed long-term studies confirming the safety of RMP as a hemostatic agent are warranted.


Subject(s)
Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/transplantation , Erythrocyte Transfusion , Erythrocytes/metabolism , Hemostasis , Hemostatic Techniques , Animals , Erythrocyte Transfusion/adverse effects , Hemostatic Techniques/adverse effects , Humans , Male , Rats, Sprague-Dawley , Time Factors , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...