Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 600
Filter
1.
Methods Mol Biol ; 2848: 59-71, 2025.
Article in English | MEDLINE | ID: mdl-39240516

ABSTRACT

Glaucoma is one of the leading causes of irreversible blindness. Stem cell therapy has shown promise in the treatment of primary open-angle glaucoma in animal models. Stem cell-free therapy using stem cell-derived trophic factors might be in demand in patients with high-risk conditions or religious restrictions. In this chapter, we describe methods for trabecular meshwork stem cell (TMSC) cultivation, secretome harvesting, and protein isolation, as well as assays to ensure the health of TMSC post-secretome harvesting and for secretome periocular injection into mice for therapeutic purposes.


Subject(s)
Stem Cells , Trabecular Meshwork , Trabecular Meshwork/metabolism , Trabecular Meshwork/cytology , Animals , Mice , Humans , Stem Cells/cytology , Stem Cells/metabolism , Regeneration , Glaucoma/therapy , Stem Cell Transplantation/methods , Secretome , Disease Models, Animal , Glaucoma, Open-Angle/therapy , Cells, Cultured , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Cell Culture Techniques/methods
2.
Stem Cell Res Ther ; 15(1): 279, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39227906

ABSTRACT

Diabetic foot ulcers (DFUs) are chronic wounds and one of the most common complications of diabetes, imposing significant physical and mental burdens on patients due to their poor prognosis and treatment efficacy. Adipose-derived stem cells (ADSCs) have been proven to promote wound healing, with studies increasingly attributing these beneficial effects to their paracrine actions. Consequently, research on ADSC secretome as a novel and promising alternative for DFU treatment has been extensively conducted. This article provides a comprehensive review of the mechanisms underlying refractory DFU wounds, the secretome of ADSCs, and its role in promoting wound healing in diabetes foot ulcers. And the review aims to provide reliable evidence for the clinical application of ADSC secretome in the treatment of refractory DFU wounds.


Subject(s)
Adipose Tissue , Diabetic Foot , Secretome , Wound Healing , Humans , Diabetic Foot/therapy , Diabetic Foot/metabolism , Diabetic Foot/pathology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Secretome/metabolism , Stem Cells/metabolism , Stem Cells/cytology , Animals
3.
Cancer Immunol Immunother ; 73(11): 229, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39249543

ABSTRACT

Immune checkpoint inhibitors are approved for recurrent/metastatic head and neck squamous cell carcinoma (HNSCC) but the response rate is only 13-18%. For an effective antitumor immune response, trafficking of immune cells to the tumor microenvironment (TME) is essential. We aimed to better understand immune cell migration as well as the involved chemokines in HNSCC. A transwell assay was used to study immune cell migration toward TME-conditioned medium. While T cell migration was not observed, conventional dendritic cell (cDC) migration was induced by TME-conditioned media. cDC migration correlated with various proteins in the TME secretome. CCL8, CXCL5, CCL13 and CCL7 were tested in validation experiments and addition of these chemokines induced cDC migration. Using single cell RNA-sequencing, we observed expression of CCL8, CXCL5, CCL13 and CCL7 in cancer-associated fibroblasts (CAFs). Depleting fibroblasts led to reduced cDC migration. Thus CAFs, while often seen as suppressors of antitumor immunity, play a role in attracting cDCs toward the head and neck cancer TME, which might be crucial for effective antitumor immunity and response to therapies. Indeed, we found RNA expression signatures of the indicated chemokines, cDC and CAF subpopulations, to be significantly higher in baseline tumor specimen of patients with a major pathological response to pre-surgical anti-PD-1 treatment compared to non-responding patients.


Subject(s)
Cell Movement , Dendritic Cells , Head and Neck Neoplasms , Tumor Microenvironment , Humans , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Tumor Microenvironment/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Secretome/metabolism , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/immunology , Chemokines/metabolism
4.
Cells ; 13(17)2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39272990

ABSTRACT

BACKGROUND: Obesity poses a significant global health challenge, given its association with the excessive accumulation of adipose tissue (AT) and various systemic disruptions. Within the adipose microenvironment, expansion and enrichment with immune cells trigger the release of inflammatory mediators and growth factors, which can disrupt tissues, including bones. While obesity's contribution to bone loss is well established, the direct impact of obese AT on osteoblast maturation remains uncertain. This study aimed to explore the influence of the secretomes from obese and lean AT on osteoblast differentiation and activity. METHODS: SAOS-2 cells were exposed to the secretomes obtained by culturing human subcutaneous AT from individuals with obesity (OATS) or lean patients, and their effects on osteoblasts were evaluated. RESULTS: In the presence of the OATS, mature osteoblasts underwent dedifferentiation, showing an increased proliferation accompanied by a morphological shift towards a mesenchymal phenotype, with detrimental effects on osteogenic markers and the calcification capacity. Concurrently, the OATS promoted the expression of mesenchymal and adipogenic markers, inducing the formation of cytoplasmic lipid droplets in SAOS-2 cells exposed to an adipogenic differentiation medium. Additionally, TGF-ß1 emerged as a key mediator of these effects, as the OATS was enriched with this growth factor. CONCLUSIONS: Our findings demonstrate that obese subcutaneous AT promotes the dedifferentiation of osteoblasts and increases the adipogenic profile in these cells.


Subject(s)
Adipogenesis , Adipose Tissue , Cell Dedifferentiation , Obesity , Osteoblasts , Phenotype , Signal Transduction , Transforming Growth Factor beta1 , Humans , Osteoblasts/metabolism , Osteoblasts/pathology , Obesity/pathology , Obesity/metabolism , Transforming Growth Factor beta1/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Secretome/metabolism , Cell Differentiation , Cell Proliferation , Osteogenesis , Male
5.
Methods Mol Biol ; 2835: 17-27, 2024.
Article in English | MEDLINE | ID: mdl-39105902

ABSTRACT

Human mesenchymal stromal cells (MSCs) have gained significant interest as cell-based therapeutics for organ restoration in the field of regenerative medicine. More recently, substantial attention has been directed toward cell-free therapy, achieved through the utilization of soluble factors possessing trophic and immunomodulatory properties present in the MSC secretome. This collection of soluble factors can be found either freely in the secretome or packed within its vesicular fraction, known as extracellular vesicles (EVs). MSCs can be derived from various tissue sources, each involving different extraction methods and yielding varying cell amounts. In this study, we describe a nonenzymatic procedure for a straightforward isolation of MSCs from the fetal dermis and the adult dermis. The results demonstrate the isolation of a cell population with a uniform MSC immunophenotype from the earliest passages (approximately 90% positive for the classical MSC markers CD90, CD105, and CD73, while negative for the hematopoietic markers CD34 and CD45, as well as HLA-DR). Additionally, we describe the procedures for cell expansion, banking, and secretome collection.


Subject(s)
Cell Separation , Dermis , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Dermis/cytology , Dermis/metabolism , Cell Separation/methods , Immunophenotyping , Cell Culture Techniques/methods , Biomarkers , Cells, Cultured , Extracellular Vesicles/metabolism , Secretome/metabolism
6.
Sci Rep ; 14(1): 19351, 2024 08 21.
Article in English | MEDLINE | ID: mdl-39169222

ABSTRACT

The most devastating feature of cancer cells is their ability to metastasize to distant sites in the body. HER2 + and TN breast cancers frequently metastasize to the brain and stay potentially dormant for years until favorable conditions support their proliferation. The sheltered and delicate nature of the brain prevents, however, early disease detection and effective delivery of therapeutic drugs. Moreover, the challenges associated with the acquisition of brain biopsies add compounding difficulties to exploring the mechanistic aspects of tumor development. To provide insights into the determinants of cancer cell behavior at the brain metastatic site, this study was aimed at exploring the early response of HER2 + breast cancer cells (SKBR3) to factors present in the brain perivascular niche. The neural microenvironment was simulated by using the secretome of a set of brain cells that come first in contact with the cancer cells upon crossing the blood brain barrier, i.e., endothelial cells, astrocytes, and microglia. Cytokine microarrays were used to investigate the secretome mediators of intercellular communication, and proteomic technologies for assessing the changes in the behavior of cancer cells upon exposure to the brain cell-secreted factors. The cytokines detected in the brain secretomes were supportive of inflammatory conditions, while the SKBR3 cells secreted numerous cancer-promoting growth factors that were either absent or present in lower abundance in the brain cell cultures, indicating that upon exposure the SKBR3 cells may have been deprived of favorable conditions for optimal growth. Altogether, the results suggest that the exposure of SKBR3 cells to the brain cell-secreted factors altered their growth potential and drove them toward a state of quiescence, with broader overall outcomes that affected cellular metabolism, adhesion and immune response processes. The findings of this study underscore the key role played by the neural niche in shaping the behavior of metastasized cancer cells, provide insights into the cellular cross-talk that may lead cancer cells into dormancy, and highlight novel opportunities for the development of metastatic breast cancer therapeutic strategies.


Subject(s)
Breast Neoplasms , Cytokines , Proteomics , Humans , Female , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Proteomics/methods , Cell Line, Tumor , Cytokines/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Tumor Microenvironment , Brain/metabolism , Brain/pathology , Secretome/metabolism , Astrocytes/metabolism , Endothelial Cells/metabolism , Proteome/metabolism , Microglia/metabolism , Receptor, ErbB-2/metabolism
7.
Cells ; 13(16)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39195236

ABSTRACT

Currently, there is a growing focus on aging and age-related diseases. The processes of aging are based on cell senescence, which results in changes in intercellular communications and pathological alterations in tissues. In the present study, we investigate the influence of senescent mesenchymal stem cells (MSCs) on endothelial cells (ECs). In order to induce senescence in MSCs, we employed a method of stress-induced senescence utilizing mitomycin C (MmC). Subsequent experiments involved the interaction of ECs with MSCs in a coculture or the treatment of ECs with the secretome of senescent MSCs. After 48 h, we assessed the EC state. Our findings revealed that direct interaction led to a decrease in EC proliferation and migratory activity of the coculture. Furthermore, there was an increase in the activity of the lysosomal compartment, as well as an upregulation of the genes P21, IL6, IL8, ITGA1, and ITGB1. Treatment of ECs with the "senescent" secretome resulted in less pronounced effects, although a decrease in proliferation and an increase in ICAM-1 expression were observed. The maintenance of high levels of typical "senescent" cytokines and growth factors after 48 h suggests that the addition of the "senescent" secretome may have a prolonged effect on the cells. It is noteworthy that in samples treated with the "senescent" secretome, the level of PDGF-AA was higher, which may explain some of the pro-regenerative effects of senescent cells. Therefore, the detected changes may underlie both the negative and positive effects of senescence. The findings provide insight into the effects of cell senescence in vitro, where many of the organism's regulatory mechanisms are absent.


Subject(s)
Cell Proliferation , Cellular Senescence , Endothelial Cells , Mesenchymal Stem Cells , Cellular Senescence/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Humans , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Endothelial Cells/cytology , Coculture Techniques , Cell Movement/drug effects , Cytokines/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Secretome/metabolism , Lysosomes/metabolism , Cells, Cultured
8.
Int J Biol Macromol ; 277(Pt 4): 134344, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39089545

ABSTRACT

Canine atopic dermatitis (AD) arises from hypersensitive immune reactions. AD symptoms entail severe pruritus and skin inflammation, with frequent relapses. Consequently, AD patients require continuous management, imposing financial burdens and mental fatigue on pet owners. In this study, we aimed to investigate the therapeutic relevance of secretome from canine adipose tissue-derived mesenchymal stem cells (MSCs), especially after encapsulation in nano-villi chitosan microspheres (CS-MS) to expect improved efficacy. Conditioned media (CM) from MSCs significantly inhibited the proliferation of splenocytes, induced the generation of regulatory T cells, and decreased mast cell degranulation. We found that beneficial soluble factors known to reduce AD symptoms, including transforming growth factor-beta 1, were detectable after sequential concentration and lyophilization of CM. The CS-MS, developed by a phase inversion regeneration method, showed high loading and sustained release of the secretome. Local injection of secretome-loaded CS-MS (ST/SC-MS) effectively reduced clinical severity compared to groups treated with secretome. Histological analysis revealed that ST/SC-MS potently suppressed epidermal hyperplasia, immunocyte infiltration and mast cell activation in the lesion. Taken together, this study presents a novel therapeutic approach exhibiting more potent and prolonged immunoregulatory efficacy of MSC secretome for canine AD treatment.


Subject(s)
Chitosan , Dermatitis, Atopic , Mesenchymal Stem Cells , Microspheres , Secretome , Dermatitis, Atopic/therapy , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Animals , Dogs , Chitosan/chemistry , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Cell Proliferation/drug effects , Mast Cells/drug effects , Mast Cells/metabolism , Mast Cells/immunology , Culture Media, Conditioned/pharmacology , Delayed-Action Preparations
9.
Mol Med ; 30(1): 132, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39187765

ABSTRACT

Cell-based therapeutic strategies have been proposed as an alternative for brain and blood vessels repair after stroke, but their clinical application is hampered by potential adverse effects. We therefore tested the hypothesis that secretome of these cells might be used instead to still focus on cell-based therapeutic strategies. We therefore characterized the composition and the effect of the secretome of brain microvascular endothelial cells (BMECs) on primary in vitro human models of angiogenesis and vascular barrier. Two different secretome batches produced in high scale (scHSP) were analysed by mass spectrometry. Human primary CD34+-derived endothelial cells (CD34+-ECs) were used as well as in vitro models of EC monolayer (CMECs) and blood-brain barrier (BBB). Cells were also exposed to oxygen-glucose deprivation (OGD) conditions and treated with scHSP during reoxygenation. Protein yield and composition of scHSP batches showed good reproducibility. scHSP increased CD34+-EC proliferation, tubulogenesis, and migration. Proteomic analysis of scHSP revealed the presence of growth factors and proteins modulating cell metabolism and inflammatory pathways. scHSP improved the integrity of CMECs, and upregulated the expression of junctional proteins. Such effects were mediated through the activation of the interferon pathway and downregulation of Wnt signalling. Furthermore, OGD altered the permeability of both CMECs and BBB, while scHSP prevented the OGD-induced vascular leakage in both models. These effects were mediated through upregulation of junctional proteins and regulation of MAPK/VEGFR2. Finally, our results highlight the possibility of using secretome from BMECs as a therapeutic alternative to promote brain angiogenesis and to protect from ischemia-induced vascular leakage.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Proteomics , Humans , Endothelial Cells/metabolism , Blood-Brain Barrier/metabolism , Proteomics/methods , Secretome/metabolism , Capillary Permeability , Brain/metabolism , Brain/blood supply , Brain/pathology , Cell Hypoxia , Proteome/metabolism , Cells, Cultured , Microvessels/metabolism , Microvessels/cytology
10.
Sci Data ; 11(1): 868, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127790

ABSTRACT

Secreted proteins regulate the balance between cellular proliferation and G0 arrest and therefore play important roles in tumour dormancy. Tumour dormancy presents a significant clinical challenge for breast cancer patients, where non-proliferating, G0-arrested cancer cells remain at metastatic sites, below the level of clinical detection, some of which can re-enter proliferation and drive tumour relapse. Knowing which secreted proteins can regulate entry into and exit from G0 allows us to manipulate their signalling to prevent tumour relapse. To identify novel secreted proteins that can promote breast cancer G0 arrest, we performed a secretome-wide, image-based screen for proteins that increase the fraction of cells in G0 arrest. From a secretome library of 1282 purified proteins, we identified 29 candidates that promote G0 arrest in non-transformed and transformed breast epithelial cells. The assay we have developed can be adapted for use in other perturbation screens in other cell types. All datasets have been made available for re-analysis and our candidate proteins are presented for alternative bioinformatic refinement or further experimental follow up.


Subject(s)
Breast Neoplasms , Humans , Breast Neoplasms/pathology , Female , Cell Cycle Checkpoints , Resting Phase, Cell Cycle , Secretome , Cell Line, Tumor
11.
Aging (Albany NY) ; 16(16): 11776-11795, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39197170

ABSTRACT

Crucial for skin homeostasis, synthesis and degradation of extracellular matrix components are orchestrated by dermal fibroblasts. During aging, alterations of component expression, such as collagens and enzymes, lead to reduction of the mechanical cutaneous tension and defects of skin wound healing. The aim of this study was to better understand the molecular alterations underwent by fibroblasts during aging by comparing secretomic and proteomic signatures of fibroblasts from young (<35years) and aged (>55years) skin donors, in quiescence or TGF-stimulated conditions, using HLPC/MS. The comparison of the secretome from young and aged fibroblasts revealed that 16 proteins in resting condition, and 11 proteins after a 24h-lasting TGF-ß1-treatment, were expressed in significant different ways between the two cell groups (fold change>2, p-value <0.05), with a 77% decrease in the number of secreted proteins in aged cells. Proteome comparison between young and aged fibroblasts identified a significant change of 63 proteins in resting condition, and 73 proteins in TGF-ß1-stimulated condition, with a 67% increase in the number of proteins in aged fibroblasts. The majority of the differentially-expressed molecules belongs to the cytoskeleton-associated proteins and aging was characterized by an increase in Coronin 1C (CORO1C), and Filamin B (FLNB) expression in fibroblasts together with a decrease in Cofilin (CFL1), and Actin alpha cardiac muscle 1 (ACTC1) detection in aged cells, these proteins being involved in actin-filament polymerization and sharing co-activity in cell motility. Our present data reinforce knowledge about an age-related alteration in the synthesis of major proteins linked to the migratory and contractile functions of dermal human fibroblasts.


Subject(s)
Aging , Cytoskeleton , Fibroblasts , Proteomics , Humans , Fibroblasts/metabolism , Cytoskeleton/metabolism , Adult , Middle Aged , Aging/metabolism , Transforming Growth Factor beta1/metabolism , Aged , Skin/metabolism , Skin/cytology , Proteome/metabolism , Cells, Cultured , Male , Secretome/metabolism , Female , Dermis/cytology , Dermis/metabolism
12.
Cell Immunol ; 403-404: 104861, 2024.
Article in English | MEDLINE | ID: mdl-39098245

ABSTRACT

The immune response to stress diverges with age, with neonatal macrophages implicated in tissue regeneration versus tissue scarring and maladaptive inflammation in adults. Integral to the macrophage stress response is the recognition of hypoxia and pathogen-associated molecular patterns (PAMPs), which are often coupled. The age-specific, cell-intrinsic nature of this stress response remains vague. To uncover age-defined divergences in macrophage crosstalk potential after exposure to hypoxia and PAMPs, we interrogated the secreted proteomes of neonatal versus adult macrophages via non-biased mass spectrometry. Through this approach, we newly identified age-specific signatures in the secretomes of neonatal versus adult macrophages in response to hypoxia and the prototypical PAMP, lipopolysaccharide (LPS). Neonatal macrophages secreted proteins most consistent with an anti-inflammatory, regenerative phenotype protective against apoptosis and oxidative stress, dependent on hypoxia inducible transcription factor-1α (HIF-1α). In contrast, adult macrophages secreted proteins consistent with a pro-inflammatory, glycolytic phenotypic signature consistent with pathogen killing. Taken together, these data uncover fundamental age and HIF-1α dependent macrophage responses that may be targeted to calibrate the innate immune response during stress and inflammation.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Lipopolysaccharides , Macrophages , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages/immunology , Macrophages/metabolism , Animals , Mice , Lipopolysaccharides/pharmacology , Lipopolysaccharides/immunology , Secretome/metabolism , Mice, Inbred C57BL , Inflammation/immunology , Inflammation/metabolism , Animals, Newborn , Immunity, Innate , Proteome/metabolism , Oxidative Stress , Cells, Cultured , Humans
13.
Parasite Immunol ; 46(7): e13056, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39073185

ABSTRACT

Co-evolutionary adaptation of hookworms with their mammalian hosts has been selected for immunoregulatory excretory/secretory (E/S) products. However, it is not known whether, or if so, how host immunological status impacts the secreted profile of hematophagous adult worms. This study interrogated the impact of host Signal transducer and activator of transcription 6 (STAT6) expression during the experimental evolution of hookworms through the sequential passage of the life cycle in either STAT6 deficient or WT C57BL/6 mice. Proteomic analysis of E/S products by LC-MS showed increased abundance of 15 proteins, including myosin-3, related to muscle function, and aconitate hydratase, related to iron homeostasis. However, most E/S proteins (174 of 337 unique identities) were decreased, including those in the Ancylostoma-secreted protein (ASP) category, and metallopeptidases. Several identified proteins are established immune-modulators such as fatty acid-binding protein homologue, cystatin, and acetylcholinesterase. Enrichment analysis of InterPro functional categories showed down-regulation of Cysteine-rich secretory proteins, Antigen 5, and Pathogenesis-related 1 proteins (CAP), Astacin-like metallopeptidase, Glycoside hydrolase, and Transthyretin-like protein groups in STAT6 KO-adapted worms. Taken together, these data indicate that in an environment lacking Type 2 immunity, hookworms alter their secretome by reducing immune evasion proteins- and increasing locomotor- and feeding-associated proteins.


Subject(s)
STAT6 Transcription Factor , Secretome , Animals , Mice , Ancylostomatoidea , Chromatography, Liquid , Helminth Proteins/metabolism , Helminth Proteins/genetics , Host-Parasite Interactions , Mice, Inbred C57BL , Mice, Knockout , Proteomics , Secretome/metabolism , STAT6 Transcription Factor/metabolism , STAT6 Transcription Factor/genetics
14.
Int J Biol Macromol ; 276(Pt 1): 133793, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38992542

ABSTRACT

Mesenchymal stem cells (MCSs) secretome provide MSC-like therapeutic effects in preclinical models of lung injury, circumventing safety concerns with the use of live cells. Secretome consists of Extracellular Vesicles (EVs), including populations of nano- to micro-sized particles (exosomes and microvesicles) delimited by a phospholipidic bilayer. However, its poor stability and bioavailability severely limit its application. The role of Hyaluronic acid (HA) as potential carrier in biomedical applications has been widely demonstrated. Here, we investigated the interplay between HA and MSCs- secretome blends and their ability to exert a bioactive effect on pulmonary differentiation in a 3D microenvironment mimicking lung niche. To this aim, the physical-chemical properties of HA/Secre blends have been characterized at low, medium and high HA Molecular Weights (MWs), by means of SEM/TEM, DLS, confocal microscopy and FTIR. Collectively physical-chemical properties highlight the interplay between the HA and the EVs. In 3D matrices, HA/Secre blends showed to promote differentiation in pulmonary lineage, improved as the MW of the HA in the blends decreased. Finally, HA/Secre blends' ability to cross an artificial mucus has been demonstrated. Overall, this work provides new insights for the development of future devices for the therapy of respiratory diseases that are still unmet.


Subject(s)
Cell Differentiation , Hyaluronic Acid , Lung , Mesenchymal Stem Cells , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Cell Differentiation/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Humans , Lung/metabolism , Lung/cytology , Secretome/metabolism , Biomimetics/methods , Cellular Microenvironment/drug effects , Extracellular Vesicles/metabolism , Extracellular Vesicles/chemistry
15.
Biochem Biophys Res Commun ; 730: 150392, 2024 Oct 20.
Article in English | MEDLINE | ID: mdl-39003867

ABSTRACT

BACKGROUND: One of the obstacles to autologous chondrocyte implantation (ACI) is obtaining a large quantity of chondrocytes without depletion of their properties. The conditioned medium (CM) from different subpopulations of stem cells (mesenchymal stromal cells (MSC) or induced pluripotent stem cells (iPSC)) could be a gamechanger. MSCs' potential is related to the donor's health and age, which could be omitted when, as a source, iPSCs are used. There is a lack of data regarding their use in the chondrocyte culture expansion. Thus, we wanted to verify whether iPSC-CM could be beneficial for the cell culture of primary chondrocyte cells. METHODS: We added the iPSC-CMs from GPCCi001-A and ND 41658*H cells to the culture of primary chondrocyte cell lines isolated from OA patients (n = 6) for other two passages. The composition of the CM was evaluated using Luminex technology. Then, we analysed the senescence, proliferation rate and using flow cytometry: viability, distribution of cell cycle phases, production of reactive oxygen species (ROS) and double-strand breaks. The cartilage-related markers were evaluated using Western blot and immunofluorescence. Additionally, a three-dimensional cell culture was used to determine the potential to form cartilage particles. RESULTS: iPSC-CM increased proliferation and diminished cell ROS production and senescence. CM influenced the cartilage-related protein expression and promoted the growth of cartilage particles. The cell exposed to CM did not lose the ECM proteins, suggesting the chondroprotective effect for prolonged culture time. CONCLUSION: Our preliminary results suggest a beneficial effect on maintaining chondrocyte biology during in vitro expansion.


Subject(s)
Cell Proliferation , Chondrocytes , Induced Pluripotent Stem Cells , Chondrocytes/metabolism , Chondrocytes/cytology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Culture Media, Conditioned/pharmacology , Culture Media, Conditioned/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Secretome/metabolism , Cell Line , Cells, Cultured , Cell Culture Techniques/methods , Reactive Oxygen Species/metabolism , Cellular Senescence
16.
Stem Cells Dev ; 33(17-18): 477-483, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39030836

ABSTRACT

There are multiple methods to prepare lipoaspirate for autologous fat transfer; however, graft retention remains unpredictable. The purpose of this study was to compare the cellular and protein composition of adipose grafts and the stromal vascular fraction (SVF) resulting from three common techniques to prepare adipose grafts. Adipose grafts were harvested from healthy donors and processed via three techniques: centrifugation (C), a single-filter (SF) device, and a double-filtration (DF) system. Part of each graft was analyzed or further processed to isolate the SVF. Cell viability, surface markers, cytokine, and growth factors were compared between the graft and SVF as well as adipose-derived stem cells (ASCs). Overall, we found variations across the three processing techniques and among the graft components (ASCs, SVF, and fat). Cell viability within the grafts was similar (94.6%, 92.3%, and 93.6%; P = 0.93). The trend was a greater percentage of ASCs from SF versus DF or centrifugation (6.95%, 4.63%, and 1.93%, respectively, P = 0.06). Adipogenic markers (adiponectin and leptin) were similar among all three grafts (P = 0.45). Markers of tissue remodeling were greatest in the SVF compared with fat and ASCs, regardless of processing technique. There was higher relative expression of MMP-9 (2×), Extracellular matrix metalloproteinase inducer (EMMPRIN) (2.5×), endoglin (5×), and IL-8 (1.5×) in the SVF (P < 0.005). Our study identified differences in cytokine expression in adipose grafts and the SVF, particularly in cytokines important in inflammation and wound healing. These secretomes may impact graft retention and fat necrosis and have the potential implications in cell-assisted lipotransfer. There were no significant differences between the final products of any of the processing techniques.


Subject(s)
Adipose Tissue , Humans , Adipose Tissue/cytology , Stromal Vascular Fraction/metabolism , Female , Adult , Cell Survival , Secretome/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Middle Aged , Cytokines/metabolism
17.
Front Immunol ; 15: 1367432, 2024.
Article in English | MEDLINE | ID: mdl-38994364

ABSTRACT

Background: Innovative therapies against bacterial infections are needed. One approach is to focus on host-directed immunotherapy (HDT), with treatments that exploit natural processes of the host immune system. The goals of this type of therapy are to stimulate protective immunity while minimizing inflammation-induced tissue damage. We use non-traditional large animal models to explore the potential of the mammosphere-derived epithelial cell (MDEC) secretome, consisting of all bioactive factors released by the cells, to modulate host immune functions. MDEC cultures are enriched for mammary stem and progenitor cells and can be generated from virtually any mammal. We previously demonstrated that the bovine MDEC secretome, collected and delivered as conditioned medium (CM), inhibits the growth of bacteria in vitro and stimulates functions related to tissue repair in cultured endothelial and epithelial cells. Methods: The immunomodulatory effects of the bovine MDEC secretome on bovine neutrophils, an innate immune cell type critical for resolving bacterial infections, were determined in vitro using functional assays. The effects of MDEC CM on neutrophil molecular pathways were explored by evaluating the production of specific cytokines by neutrophils and examining global gene expression patterns in MDEC CM-treated neutrophils. Enzyme linked immunosorbent assays were used to determine the concentrations of select proteins in MDEC CM and siRNAs were used to reduce the expression of specific MDEC-secreted proteins, allowing for the identification of bioactive factors modulating neutrophil functions. Results: Neutrophils exposed to MDEC secretome exhibited increased chemotaxis and phagocytosis and decreased intracellular reactive oxygen species and extracellular trap formation, when compared to neutrophils exposed to control medium. C-X-C motif chemokine 6, superoxide dismutase, peroxiredoxin-2, and catalase, each present in the bovine MDEC secretome, were found to modulate neutrophil functions. Conclusion: The MDEC secretome administered to treat bacterial infections may increase neutrophil recruitment to the site of infection, stimulate pathogen phagocytosis by neutrophils, and reduce neutrophil-produced ROS accumulation. As a result, pathogen clearance might be improved and local inflammation and tissue damage reduced.


Subject(s)
Epithelial Cells , Neutrophils , Secretome , Animals , Cattle , Neutrophils/immunology , Neutrophils/metabolism , Epithelial Cells/metabolism , Epithelial Cells/immunology , Secretome/metabolism , Female , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Phagocytosis , Mammary Glands, Animal/immunology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/cytology , Cells, Cultured , Reactive Oxygen Species/metabolism
18.
Cancer Rep (Hoboken) ; 7(7): e2080, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38967113

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is a malignant brain tumor that frequently occurs alongside other central nervous system (CNS) conditions. The secretome of GBM cells contains a diverse array of proteins released into the extracellular space, influencing the tumor microenvironment. These proteins can serve as potential biomarkers for GBM due to their involvement in key biological processes, exploring the secretome biomarkers in GBM research represents a cutting-edge strategy with significant potential for advancing diagnostic precision, treatment monitoring, and ultimately improving outcomes for patients with this challenging brain cancer. AIM: This study was aimed to investigate the roles of secretome biomarkers and their pathwayes in GBM through bioinformatics analysis. METHODS AND RESULTS: Using data from the Gene Expression Omnibus and the Cancer Genome Atlas datasets-where both healthy and cancerous samples were analyzed-we used a quantitative analytical framework to identify differentially expressed genes (DEGs) and cell signaling pathways that might be related to GBM. Then, we performed gene ontology studies and hub protein identifications to estimate the roles of these DEGs after finding disease-gene connection networks and signaling pathways. Using the GEPIA Proportional Hazard Model and the Kaplan-Meier estimator, we widened our analysis to identify the important genes that may play a role in both progression and the survival of patients with GBM. In total, 890 DEGs, including 475 and 415 upregulated and downregulated were identified, respectively. Our results revealed that SQLE, DHCR7, delta-1 phospholipase C (PLCD1), and MINPP1 genes are highly expressed, and the Enolase 2 (ENO2) and hexokinase-1 (HK1) genes are low expressions. CONCLUSION: Hence, our findings suggest novel mechanisms that affect the occurrence of GBM development, growth, and/or establishment and may also serve as secretory biomarkers for GBM prognosis and possible targets for therapy. So, continued research in this field may uncover new avenues for therapeutic interventions and contribute to the ongoing efforts to combat GBM effectively.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms , Computational Biology , Gene Expression Regulation, Neoplastic , Glioblastoma , Neoplastic Stem Cells , Humans , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/mortality , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Secretome/metabolism , Gene Expression Profiling , Signal Transduction , Prognosis , Gene Regulatory Networks , Protein Interaction Maps , Tumor Microenvironment
19.
Stem Cell Res Ther ; 15(1): 230, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075600

ABSTRACT

BACKGROUND: Radiation therapy is the standard of care for central nervous system tumours. Despite the success of radiation therapy in reducing tumour mass, irradiation (IR)-induced vasculopathies and neuroinflammation contribute to late-delayed complications, neurodegeneration, and premature ageing in long-term cancer survivors. Mesenchymal stromal cells (MSCs) are adult stem cells that facilitate tissue integrity, homeostasis, and repair. Here, we investigated the potential of the iPSC-derived MSC (iMSC) secretome in immunomodulation and vasculature repair in response to radiation injury utilizing human cell lines. METHODS: We generated iPSC-derived iMSC lines and evaluated the potential of their conditioned media (iMSC CM) to treat IR-induced injuries in human monocytes (THP1) and brain vascular endothelial cells (hCMEC/D3). We further assessed factors in the iMSC secretome, their modulation, and the molecular pathways they elicit. RESULTS: Increasing doses of IR disturbed endothelial tube and spheroid formation in hCMEC/D3. When IR-injured hCMEC/D3 (IR ≤ 5 Gy) were treated with iMSC CM, endothelial cell viability, adherence, spheroid compactness, and proangiogenic sprout formation were significantly ameliorated, and IR-induced ROS levels were reduced. iMSC CM augmented tube formation in cocultures of hCMEC/D3 and iMSCs. Consistently, iMSC CM facilitated angiogenesis in a zebrafish model in vivo. Furthermore, iMSC CM suppressed IR-induced NFκB activation, TNF-α release, and ROS production in THP1 cells. Additionally, iMSC CM diminished NF-kB activation in THP1 cells cocultured with irradiated hCMEC/D3, iMSCs, or HMC3 microglial lines. The cytokine array revealed that iMSC CM contains the proangiogenic and immunosuppressive factors MCP1/CCL2, IL6, IL8/CXCL8, ANG (Angiogenin), GROα/CXCL1, and RANTES/CCL5. Common promoter regulatory elements were enriched in TF-binding motifs such as androgen receptor (ANDR) and GATA2. hCMEC/D3 phosphokinome profiling revealed increased expression of pro-survival factors, the PI3K/AKT/mTOR modulator PRAS40 and ß-catenin in response to CM. The transcriptome analysis revealed increased expression of GATA2 in iMSCs and the enrichment of pathways involved in RNA metabolism, translation, mitochondrial respiration, DNA damage repair, and neurodevelopment. CONCLUSIONS: The iMSC secretome is a comodulated composite of proangiogenic and immunosuppressive factors that has the potential to alleviate radiation-induced vascular endothelial cell damage and immune activation.


Subject(s)
Endothelial Cells , Induced Pluripotent Stem Cells , Mesenchymal Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Secretome/metabolism , Animals , Zebrafish , Culture Media, Conditioned/pharmacology , Neovascularization, Physiologic/radiation effects
20.
Hum Cell ; 37(5): 1243-1275, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38909330

ABSTRACT

The currently available osteoarthritis (OA) treatments offer symptoms' relief without disease-modifying effects. Increasing evidence supports the role of human mesenchymal stem cells (MSCs) to drive beneficial effects provided by their secretome and extracellular vesicles (EVs), which includes trophic and biologically active factors. Aim of this study was to evaluate the in vitro literature to understand the potential of human secretome and EVs for OA treatment and identify trends, gaps, and potential translational challenges. A systematic review was performed on PubMed, Embase, and Web-of-Science, identifying 58 studies. The effects of secretome and EVs were analysed on osteoarthritic cells regarding anabolic, anti-apoptotic/anti-inflammatory and catabolic/pro-inflammatory/degenerative activity, chondroinduction, and immunomodulation. The results showed that MSC-derived EVs elicit an increase in proliferation and migration, reduction of cell death and inflammation, downregulation of catabolic pathways, regulation of immunomodulation, and promotion of anabolic processes in arthritic cells. However, a high heterogeneity in several technical or more applicative aspects emerged. In conclusion, the use of human secretome and EVs as strategy to address OA processes has overall positive effects and disease-modifying potential. However, it is crucial to reduce protocol variability and strive toward a higher standardization, which will be essential for the translation of this promising OA treatment from the in vitro research setting to the clinical practice.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Osteoarthritis , Secretome , Humans , Osteoarthritis/therapy , Osteoarthritis/metabolism , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Secretome/metabolism , Immunomodulation , Cell Proliferation , Cell Movement
SELECTION OF CITATIONS
SEARCH DETAIL