Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 568
Filtrer
1.
Nat Commun ; 15(1): 4862, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862464

RÉSUMÉ

As spaceflight becomes more common with commercial crews, blood-based measures of crew health can guide both astronaut biomedicine and countermeasures. By profiling plasma proteins, metabolites, and extracellular vesicles/particles (EVPs) from the SpaceX Inspiration4 crew, we generated "spaceflight secretome profiles," which showed significant differences in coagulation, oxidative stress, and brain-enriched proteins. While >93% of differentially abundant proteins (DAPs) in vesicles and metabolites recovered within six months, the majority (73%) of plasma DAPs were still perturbed post-flight. Moreover, these proteomic alterations correlated better with peripheral blood mononuclear cells than whole blood, suggesting that immune cells contribute more DAPs than erythrocytes. Finally, to discern possible mechanisms leading to brain-enriched protein detection and blood-brain barrier (BBB) disruption, we examined protein changes in dissected brains of spaceflight mice, which showed increases in PECAM-1, a marker of BBB integrity. These data highlight how even short-duration spaceflight can disrupt human and murine physiology and identify spaceflight biomarkers that can guide countermeasure development.


Sujet(s)
Coagulation sanguine , Barrière hémato-encéphalique , Encéphale , Homéostasie , Stress oxydatif , Vol spatial , Animaux , Humains , Encéphale/métabolisme , Barrière hémato-encéphalique/métabolisme , Souris , Coagulation sanguine/physiologie , Mâle , Sécrétome/métabolisme , Souris de lignée C57BL , Vésicules extracellulaires/métabolisme , Protéomique/méthodes , Marqueurs biologiques/métabolisme , Marqueurs biologiques/sang , Femelle , Adulte , Protéines du sang/métabolisme , Adulte d'âge moyen , Agranulocytes/métabolisme , Protéome/métabolisme
2.
J Med Microbiol ; 73(6)2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38836745

RÉSUMÉ

Introduction. The fungal pathogen Aspergillus fumigatus can induce prolonged colonization of the lungs of susceptible patients, resulting in conditions such as allergic bronchopulmonary aspergillosis and chronic pulmonary aspergillosis.Hypothesis. Analysis of the A. fumigatus secretome released during sub-lethal infection of G. mellonella larvae may give an insight into products released during prolonged human colonisation.Methodology. Galleria mellonella larvae were infected with A. fumigatus, and the metabolism of host carbohydrate and proteins and production of fungal virulence factors were analysed. Label-free qualitative proteomic analysis was performed to identify fungal proteins in larvae at 96 hours post-infection and also to identify changes in the Galleria proteome as a result of infection.Results. Infected larvae demonstrated increasing concentrations of gliotoxin and siderophore and displayed reduced amounts of haemolymph carbohydrate and protein. Fungal proteins (399) were detected by qualitative proteomic analysis in cell-free haemolymph at 96 hours and could be categorized into seven groups, including virulence (n = 25), stress response (n = 34), DNA repair and replication (n = 39), translation (n = 22), metabolism (n = 42), released intracellular (n = 28) and cellular development and cell cycle (n = 53). Analysis of the Gallerial proteome at 96 hours post-infection revealed changes in the abundance of proteins associated with immune function, metabolism, cellular structure, insect development, transcription/translation and detoxification.Conclusion. Characterizing the impact of the fungal secretome on the host may provide an insight into how A. fumigatus damages tissue and suppresses the immune response during long-term pulmonary colonization.


Sujet(s)
Aspergillus fumigatus , Protéines fongiques , Larve , Papillons de nuit , Animaux , Aspergillus fumigatus/métabolisme , Larve/microbiologie , Papillons de nuit/microbiologie , Protéines fongiques/métabolisme , Protéines fongiques/génétique , Sécrétome/métabolisme , Protéomique , Facteurs de virulence/métabolisme , Protéome/analyse , Hémolymphe/microbiologie , Hémolymphe/métabolisme , Virulence , Aspergillose/microbiologie , Aspergillose/métabolisme
3.
Cells ; 13(11)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38891113

RÉSUMÉ

Tigilanol tiglate (TT, also known as EBC-46) is a novel, plant-derived diterpene ester possessing anticancer and wound-healing properties. Here, we show that TT-evoked PKC-dependent S985 phosphorylation of the tyrosine kinase MET leads to subsequent degradation of tyrosine phosphorylated p-Y1003 and p-Y1234/5 MET species. PKC inhibition with BIM-1 blocked S985 phosphorylation of MET and led to MET cell surface accumulation. Treatment with metalloproteinase inhibitors prevented MET-ECD release into cell culture media, which was also blocked by PKC inhibitors. Furthermore, unbiased secretome analysis, performed using TMT-technology, identified additional targets of TT-dependent release of cell surface proteins from H357 head and neck cancer cells. We confirm that the MET co-signalling receptor syndecan-1 was cleaved from the cell surface in response to TT treatment. This was accompanied by rapid cleavage of the cellular junction adhesion protein Nectin-1 and the nerve growth factor receptor NGFRp75/TNFR16. These findings, that TT is a novel negative regulator of protumorigenic c-MET and NGFRp75/TNFR16 signalling, as well as regulating Nectin-1-mediated cell adhesion, further contribute to our understanding of the mode of action and efficacy of TT in the treatment of solid tumours.


Sujet(s)
Tumeurs de la tête et du cou , Protéines proto-oncogènes c-met , Humains , Protéines proto-oncogènes c-met/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/génétique , Lignée cellulaire tumorale , Sécrétome/métabolisme , Diterpènes/pharmacologie , Protéines membranaires/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Syndécane-1/métabolisme , Nectines/métabolisme , Protéine kinase C/métabolisme
4.
Stem Cell Res Ther ; 15(1): 137, 2024 May 12.
Article de Anglais | MEDLINE | ID: mdl-38735979

RÉSUMÉ

Scar tissue is the inevitable result of repairing human skin after it has been subjected to external destructive stimuli. It leads to localized damage to the appearance of the skin, accompanied by symptoms such as itching and pain, which reduces the quality of life of the patient and causes serious medical burdens. With the continuous development of economy and society, there is an increasing demand for beauty. People are looking forward to a safer and more effective method to eliminate pathological scarring. In recent years, adipose-derived stem cells (ADSCs) have received increasing attention from researchers. It can effectively improve pathological scarring by mediating inflammation, regulating fibroblast proliferation and activation, and vascular reconstruction. This review focuses on the pathophysiological mechanisms of hypertrophic scarring, summarizing the therapeutic effects of in vitro, in vivo, and clinical studies on the therapeutic effects of ADSCs in the field of hypertrophic scarring prevention and treatment, the latest application techniques, such as cell-free therapies utilizing ADSCs, and discussing the advantages and limitations of ADSCs. Through this review, we hope to further understand the characterization of ADSC and clarify the effectiveness of its application in hypertrophic scarring treatment, so as to provide clinical guidance.


Sujet(s)
Tissu adipeux , Cicatrice hypertrophique , Humains , Cicatrice hypertrophique/thérapie , Cicatrice hypertrophique/métabolisme , Cicatrice hypertrophique/anatomopathologie , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Cellules souches/métabolisme , Cellules souches/cytologie , Sécrétome/métabolisme , Animaux , Transplantation de cellules souches/méthodes
5.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-38731966

RÉSUMÉ

Leukemias are among the most prevalent types of cancer worldwide. Bone marrow mesenchymal stem cells (MSCs) participate in the development of a suitable niche for hematopoietic stem cells, and are involved in the development of diseases such as leukemias, to a yet unknown extent. Here we described the effect of secretome of bone marrow MSCs obtained from healthy donors and from patients with acute myeloid leukemia (AML) on leukemic cell lineages, sensitive (K562) or resistant (K562-Lucena) to chemotherapy drugs. Cell proliferation, viability and death were evaluated, together with cell cycle, cytokine production and gene expression of ABC transporters and cyclins. The secretome of healthy MSCs decreased proliferation and viability of both K562 and K562-Lucena cells; moreover, an increase in apoptosis and necrosis rates was observed, together with the activation of caspase 3/7, cell cycle arrest in G0/G1 phase and changes in expression of several ABC proteins and cyclins D1 and D2. These effects were not observed using the secretome of MSCs derived from AML patients. In conclusion, the secretome of healthy MSCs have the capacity to inhibit the development of leukemia cells, at least in the studied conditions. However, MSCs from AML patients seem to have lost this capacity, and could therefore contribute to the development of leukemia.


Sujet(s)
Prolifération cellulaire , Leucémie aigüe myéloïde , Cellules souches mésenchymateuses , Humains , Cellules souches mésenchymateuses/métabolisme , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/génétique , Cellules K562 , Apoptose , Sécrétome/métabolisme , Adulte d'âge moyen , Femelle , Mâle , Cellules de la moelle osseuse/métabolisme , Lignage cellulaire/génétique , Survie cellulaire , Adulte
6.
Sci Rep ; 14(1): 10243, 2024 05 03.
Article de Anglais | MEDLINE | ID: mdl-38702388

RÉSUMÉ

The widespread use of multipotent mesenchymal stromal cell-derived secretome (MSC-sec) requires optimal preservation methods. Lyophilization offers benefits like concentrating the secretome, reducing the storage volume, and making storage conditions more flexible. This study evaluated the influence of storage duration and temperature on lyophilized MSC-sec. The conditioned medium from Wharton's jelly MSCs was stored at - 80 °C or lyophilized with or without trehalose. Lyophilized formulations were kept at - 80 °C, - 20 °C, 4 °C, or room temperature (RT) for 3 and 30 months. After storage and reconstitution, the levels of growth factors and cytokines were assessed using multiplex assay. The storage of lyophilized MSC-sec at - 80 °C ensured biomolecule preservation for 3 and 30 months. Following 3 month storage at 4 °C and RT, a notable decrease occurred in BDNF, bNGF, and sVCAM-1 levels. Prolonged 30 month storage at the same temperatures significantly reduced BDNF, bNGF, VEGF-A, IL-6, and sVCAM-1, while storage at - 20 °C decreased BDNF, bNGF, and VEGF- A levels. Trehalose supplementation of MSC-sec improved the outcome during storage at 4 °C and RT. Proper storage conditions were crucial for the preservation of lyophilized MSC-sec composition. Short-term storage at various temperatures maintained over 60% of the studied growth factors and cytokines; long-term preservation was only adequate at -80 °C.


Sujet(s)
Lyophilisation , Cellules souches mésenchymateuses , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Humains , Sécrétome/métabolisme , Tréhalose/métabolisme , Tréhalose/pharmacologie , Cytokines/métabolisme , Cellules cultivées , Milieux de culture conditionnés/composition chimique , Cryoconservation/méthodes , Température
7.
Asian Pac J Cancer Prev ; 25(5): 1803-1813, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38809653

RÉSUMÉ

BACKGROUND: Breast cancer stem cells (BCSCs) play a role in the high rates of resistance, recurrence, and metastasis. The precise biomarkers of BCSCs can assist effectively in identifying cancer, assessing prognosis, diagnosing, and monitoring therapy. The aim of this study was to give a complete analysis for predicting specific biomarkers of BCSCs. METHODS: We aggregated profile datasets in this work to shed light on the underlying critical genes and pathways of BCSCs. We obtained two expression profiling by array datasets (GSE7513 and GSE7515) from the Gene Expression Omnibus (GEO) database to identify biomarkers in BCSCs. Enrichr was used to do functional analysis, including gene ontology (GO) and reactome pathway. Furthermore, the protein-protein interaction (PPI) of these differential expression genes (DEGs) was visualized using Cytoscape with the search tool for the retrieval of interacting genes (STRING). The hub genes in the PPI network were chosen for further investigation. RESULTS: We identified 65 up-regulated and 190 down- regulated DEGs and the GO enrichment analysis revealed that these DEGs were enriched in biological process related to tumorigenesis and stemness, including alter the extracellular matrix's physicochemical properties, cytoskeletal reorganisation, adhesion, motility, migration, growth, and survival. The Reactome analysis indicated that these DEGs were also involved in modulating function of ECM, regulation cancer metabolism and angiogenesis, tumor growth, proliferation, and metastasis. CONCLUSION: Our bioinformatic study revealed that FYN, INADL, OCLN, F11R, and TOP2A were potential biomarker panel of BCSCs from secretome.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du sein , Cellules souches tumorales , Cartes d'interactions protéiques , Humains , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Femelle , Sécrétome/métabolisme , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Réseaux de régulation génique , Biologie informatique/méthodes , Pronostic
8.
EBioMedicine ; 103: 105145, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38713924

RÉSUMÉ

BACKGROUND: There is increased evidence that the effects of stem cells can mostly be duplicated by administration of their secretome which might streamline the translation towards the clinics. METHODS: The 12-patient SECRET-HF phase 1 trial has thus been designed to determine the feasibility and safety of repeated intravenous injections of the extracellular vesicle (EV)-enriched secretome of cardiovascular progenitor cells differentiated from pluripotent stem cells in severely symptomatic patients with drug-refractory left ventricular (LV) dysfunction secondary to non-ischemic dilated cardiomyopathy. Here we report the case of the first treated patient (baseline NYHA class III; LV Ejection Fraction:25%) in whom a dose of 20 × 109 particles/kg was intravenously infused three times three weeks apart. FINDINGS: In addition to demonstrating the feasibility of producing a cardiac cell secretome compliant with Good Manufacturing Practice standards, this case documents the excellent tolerance of its repeated delivery, without any adverse events during or after infusions. Six months after the procedure, the patient is in NYHA Class II with improved echo parameters, a reduced daily need for diuretics (from 240 mg to 160 mg), no firing from the previously implanted automatic internal defibrillator and no alloimmunization against the drug product, thereby supporting its lack of immunogenicity. INTERPRETATION: The rationale underlying the intravenous route is that the infused EV-enriched secretome may act by rewiring endogenous immune cells, both circulating and in peripheral organs, to take on a reparative phenotype. These EV-modified immune cells could then traffic to the heart to effect tissue repair, including mitigation of inflammation which is a hallmark of cardiac failure. FUNDING: This trial is funded by the French Ministry of Health (Programme Hospitalier de Recherche CliniqueAOM19330) and the "France 2030" National Strategy Program (ANR-20-F2II-0003). It is sponsored by Assistance Publique-Hôpitaux de Paris.


Sujet(s)
Défaillance cardiaque , Sécrétome , Humains , Défaillance cardiaque/thérapie , Défaillance cardiaque/métabolisme , Défaillance cardiaque/étiologie , Sécrétome/métabolisme , Mâle , Vésicules extracellulaires/métabolisme , Adulte d'âge moyen , Résultat thérapeutique
9.
Talanta ; 276: 126216, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38761653

RÉSUMÉ

Human amniotic mesenchymal stromal cells (hAMSCs) have unique immunomodulatory properties making them attractive candidates for regenerative applications in inflammatory diseases. Most of their beneficial properties are mediated through their secretome. The bioactive factors concurring to its therapeutic activity are still unknown. Evidence suggests synergy between the two main components of the secretome, soluble factors and vesicular fractions, pivotal in shifting inflammation and promoting self-healing. Biological variability and the absence of quality control (QC) protocols hinder secretome-based therapy translation to clinical applications. Moreover, vesicular secretome contains a multitude of particles with varying size, cargos and functions whose complexity hinders full characterization and comprehension. This study achieved a significant advancement in secretome characterization by utilizing native, FFF-based separation and characterizing extracellular vesicles derived from hAMSCs. This was accomplished by obtaining dimensionally homogeneous fractions then characterized based on their protein content, potentially enabling the identification of subpopulations with diverse functionalities. This method proved to be successful as an independent technique for secretome profiling, with the potential to contribute to the standardization of a qualitative method. Additionally, it served as a preparative separation tool, streamlining populations before ELISA and LC-MS characterization. This approach facilitated the categorization of distinctive and recurring proteins, along with the identification of clusters associated with vesicle activity and functions. However, the presence of proteins unique to each fraction obtained through the FFF separation tool presents a challenge for further analysis of the protein content within these cargoes.


Sujet(s)
Amnios , Vésicules extracellulaires , Cellules souches mésenchymateuses , Sécrétome , Humains , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Sécrétome/métabolisme , Amnios/composition chimique , Amnios/cytologie , Amnios/métabolisme , Vésicules extracellulaires/composition chimique , Vésicules extracellulaires/métabolisme , Contrôle de qualité , Cellules cultivées
10.
J Proteome Res ; 23(6): 2160-2168, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38767394

RÉSUMÉ

Resistance is a major problem with effective cancer treatment and the stroma forms a significant portion of the tumor mass but traditional drug screens involve cancer cells alone. Cancer-associated fibroblasts (CAFs) are a major tumor stroma component and its secreted proteins may influence the function of cancer cells. The majority of secretome studies compare different cancer or CAF cell lines exclusively. Here, we present the direct characterization of the secreted protein profiles between CAFs and KRAS mutant-cancer cell lines from colorectal, lung, and pancreatic tissues using multiplexed mass spectrometry. 2573 secreted proteins were annotated, and differential analysis highlighted understudied CAF-enriched secreted proteins, including Wnt family member 5B (WNT5B), in addition to established CAF markers, such as collagens. The functional role of CAF secreted proteins was explored by assessing its effect on the response to 97 anticancer drugs since stromal cells may cause a differing cancer drug response, which may be missed on routine drug screening using cancer cells alone. CAF secreted proteins caused specific effects on each of the cancer cell lines, which highlights the complexity and challenges in cancer treatment and so the importance to consider stromal elements.


Sujet(s)
Fibroblastes associés au cancer , Sécrétome , Humains , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/anatomopathologie , Lignée cellulaire tumorale , Sécrétome/métabolisme , Antinéoplasiques/pharmacologie , Résistance aux médicaments antinéoplasiques , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Spectrométrie de masse , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Protéomique/méthodes , Tumeurs du poumon/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique
11.
PLoS One ; 19(5): e0292978, 2024.
Article de Anglais | MEDLINE | ID: mdl-38728307

RÉSUMÉ

Endosalpingiosis (ES) and endometriosis (EM) refer to the growth of tubal and endometrial epithelium respectively, outside of their site of origin. We hypothesize that uterine secretome factors drive ectopic growth. To test this, we developed a mouse model of ES and EM using tdTomato (tdT) transgenic fluorescent mice as donors. To block implantation factors, progesterone knockout (PKO) tdT mice were created. Fluorescent lesions were present after oviduct implantation with and without WT endometrium. Implantation was increased (p<0.05) when tdt oviductal tissue was implanted with endometrium compared to oviductal tissue alone. Implantation was reduced (p<0.0005) in animals implanted with minced tdT oviductal tissue with PKO tdT endometrium compared to WT endometrium. Finally, oviductal tissues was incubated with and without a known implantation factor, leukemia inhibitory factor (LIF) prior to and during implantation. LIF promoted lesion implantation. In conclusion, endometrial derived implantation factors, such as LIF, are necessary to initiate ectopic tissue growth. We have developed an animal model of ectopic growth of gynecologic tissues in a WT mouse which will potentially allow for development of new prevention and treatment modalities.


Sujet(s)
Endométriose , Endomètre , Utérus , Animaux , Femelle , Souris , Endométriose/métabolisme , Endométriose/anatomopathologie , Endométriose/génétique , Utérus/métabolisme , Endomètre/métabolisme , Facteur inhibiteur de la leucémie/métabolisme , Facteur inhibiteur de la leucémie/génétique , Sécrétome/métabolisme , Souris transgéniques , Modèles animaux de maladie humaine , Trompes utérines/métabolisme , Progestérone/métabolisme , Souris knockout , Implantation embryonnaire/physiologie
12.
Stem Cell Res Ther ; 15(1): 119, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38659070

RÉSUMÉ

BACKGROUND: Adipose stromal cells (ASC) are a form of mesenchymal stromal cells that elicit effects primarily via secreted factors, which may have advantages for the treatment of injury or disease. Several previous studies have demonstrated a protective role for MSC/ASC on mitigating acute kidney injury but whether ASC derived factors could hasten recovery from established injury has not been evaluated. METHODS: We generated a concentrated secretome (CS) of human ASC under well-defined conditions and evaluated its ability to improve the recovery of renal function in a preclinical model of acute kidney injury (AKI) in rats. 24 h following bilateral ischemia/reperfusion (I/R), rats were randomized following determination of plasma creatinine into groups receiving vehicle -control or ASC-CS treatment by subcutaneous injection (2 mg protein/kg) and monitored for evaluation of renal function, structure and inflammation. RESULTS: Renal function, assessed by plasma creatinine levels, recovered faster in ASC-CS treated rats vs vehicle. The most prominent difference between the ASC-CS treated vs vehicle was observed in rats with the most severe degree of initial injury (Pcr > 3.0 mg/dl 24 h post I/R), whereas rats with less severe injury (Pcr < 2.9 mg/dl) recovered quickly regardless of treatment. The quicker recovery of ASC-treated rats with severe injury was associated with less tissue damage, inflammation, and lower plasma angiopoietin 2. In vitro, ASC-CS attenuated the activation of the Th17 phenotype in lymphocytes isolated from injured kidneys. CONCLUSIONS: Taken together, these data suggest that ASC-CS represents a potent therapeutic option to improve established AKI.


Sujet(s)
Atteinte rénale aigüe , Inflammation , Atteinte rénale aigüe/thérapie , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Animaux , Rats , Humains , Inflammation/anatomopathologie , Inflammation/métabolisme , Mâle , Sécrétome/métabolisme , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Rat Sprague-Dawley , Injections sous-cutanées , Rein/métabolisme , Rein/anatomopathologie , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/thérapie , Cellules stromales/métabolisme
13.
Front Immunol ; 15: 1232070, 2024.
Article de Anglais | MEDLINE | ID: mdl-38638443

RÉSUMÉ

Chronic liver diseases, such as non-alcoholic steatohepatitis (NASH)-induced cirrhosis, are characterized by an increasing accumulation of stressed, damaged, or dying hepatocytes. Hepatocyte damage triggers the activation of resident immune cells, such as Kupffer cells (KC), as well as the recruitment of immune cells from the circulation toward areas of inflammation. After infiltration, monocytes differentiate into monocyte-derived macrophages (MoMF) which are functionally distinct from resident KC. We herein aim to compare the in vitro signatures of polarized macrophages and activated hepatic stellate cells (HSC) with ex vivo-derived disease signatures from human NASH. Furthermore, to shed more light on HSC activation and liver fibrosis progression, we investigate the effects of the secretome from primary human monocytes, macrophages, and NK cells on HSC activation. Interleukin (IL)-4 and IL-13 treatment induced transforming growth factor beta 1 (TGF-ß1) secretion by macrophages. However, the supernatant transfer did not induce HSC activation. Interestingly, PMA-activated macrophages showed strong induction of the fibrosis response genes COL10A1 and CTGF, while the supernatant of IL-4/IL-13-treated monocytes induced the upregulation of COL3A1 in HSC. The supernatant of PMA-activated NK cells had the strongest effect on COL10A1 induction in HSC, while IL-15-stimulated NK cells reduced the expression of COL1A1 and CTGF. These data indicate that other factors, aside from the well-known cytokines and chemokines, might potentially be stronger contributors to the activation of HSCs and induction of a fibrotic response, indicating a more diverse and complex role of monocytes, macrophages, and NK cells in liver fibrosis progression.


Sujet(s)
Cellules de Küpffer , Stéatose hépatique non alcoolique , Humains , Cellules de Küpffer/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Interleukine-13/métabolisme , Sécrétome , Macrophages , Cirrhose du foie , Cellules tueuses naturelles/métabolisme
14.
Mol Biol Rep ; 51(1): 570, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38658405

RÉSUMÉ

INTRODUCTION: Spinal cord injury (SCI) leads to significant destruction of nerve tissue, causing the degeneration of axons and the formation of cystic cavities. This study aimed to examine the characteristics of human umbilical cord-derived mesenchymal stem cells (HUCMSCs) cultured in a serum-free conditioned medium (CM) and assess their effectiveness in a well-established hemitransection SCI model. MATERIALS AND METHODS: In this study, HUCMSCs cultured medium was collected and characterized by measuring IL-10 and identifying proteomics using mass spectroscopy. This collected serum-free CM was further used in the experiments to culture and characterize the HUMSCs. Later, neuronal cells derived from CM-enriched HUCMSC were tested sequentially using an injectable caffeic acid-bioconjugated gelatin (CBG), which was further transplanted in a hemitransection SCI model. In vitro, characterization of CM-enriched HUCMSCs and differentiated neuronal cells was performed using flow cytometry, immunofluorescence, electron microscopy, and post-transplant analysis using immunohistology analysis, qPCR, in vivo bioluminescence imaging, and behavioral analysis using an infrared actimeter. RESULTS: The cells that were cultured in the conditioned media produced a pro-inflammatory cytokine called IL-10. Upon examining the secretome of the conditioned media, the Kruppel-like family of KRAB and zinc-finger proteins (C2H2 and C4) were found to be activated. Transcriptome analysis also revealed an increased expression of ELK-1, HOXD8, OTX2, YY1, STAT1, ETV7, and PATZ1 in the conditioned media. Furthermore, the expression of Human Stem-101 confirmed proliferation during the first 3 weeks after transplantation, along with the migration of CBG-UCNSC cells within the transplanted area. The gene analysis showed increased expression of Nestin, NeuN, Calb-2, Msi1, and Msi2. The group that received CBG-UCNSC therapy showed a smooth recovery by the end of week 2, with most rats regaining their walking abilities similar to those before the spinal cord injury by week 5. CONCLUSIONS: In conclusion, the CBG-UCNSC method effectively preserved the integrity of the transplanted neuronal-like cells and improved locomotor function. Thus, CM-enriched cells can potentially reduce biosafety risks associated with animal content, making them a promising option for clinical applications in treating spinal cord injuries.


Sujet(s)
Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Traumatismes de la moelle épinière , Transcriptome , Cordon ombilical , Traumatismes de la moelle épinière/thérapie , Traumatismes de la moelle épinière/métabolisme , Traumatismes de la moelle épinière/génétique , Cellules souches mésenchymateuses/métabolisme , Milieux de culture conditionnés/pharmacologie , Cordon ombilical/cytologie , Cordon ombilical/métabolisme , Humains , Animaux , Transplantation de cellules souches mésenchymateuses/méthodes , Transcriptome/génétique , Rats , Sécrétome/métabolisme , Différenciation cellulaire , Neurones/métabolisme , Modèles animaux de maladie humaine , Interleukine-10/génétique , Interleukine-10/métabolisme , Cellules cultivées , Protéomique/méthodes
15.
BMC Plant Biol ; 24(1): 251, 2024 Apr 06.
Article de Anglais | MEDLINE | ID: mdl-38582844

RÉSUMÉ

BACKGROUND: Many parasitic plants of the genera Striga and Cuscuta inflict huge agricultural damage worldwide. To form and maintain a connection with a host plant, parasitic plants deploy virulence factors (VFs) that interact with host biology. They possess a secretome that represents the complement of proteins secreted from cells and like other plant parasites such as fungi, bacteria or nematodes, some secreted proteins represent VFs crucial to successful host colonisation. Understanding the genome-wide complement of putative secreted proteins from parasitic plants, and their expression during host invasion, will advance understanding of virulence mechanisms used by parasitic plants to suppress/evade host immune responses and to establish and maintain a parasite-host interaction. RESULTS: We conducted a comparative analysis of the secretomes of root (Striga spp.) and shoot (Cuscuta spp.) parasitic plants, to enable prediction of candidate VFs. Using orthogroup clustering and protein domain analyses we identified gene families/functional annotations common to both Striga and Cuscuta species that were not present in their closest non-parasitic relatives (e.g. strictosidine synthase like enzymes), or specific to either the Striga or Cuscuta secretomes. For example, Striga secretomes were strongly associated with 'PAR1' protein domains. These were rare in the Cuscuta secretomes but an abundance of 'GMC oxidoreductase' domains were found, that were not present in the Striga secretomes. We then conducted transcriptional profiling of genes encoding putatively secreted proteins for the most agriculturally damaging root parasitic weed of cereals, S. hermonthica. A significant portion of the Striga-specific secretome set was differentially expressed during parasitism, which we probed further to identify genes following a 'wave-like' expression pattern peaking in the early penetration stage of infection. We identified 39 genes encoding putative VFs with functions such as cell wall modification, immune suppression, protease, kinase, or peroxidase activities, that are excellent candidates for future functional studies. CONCLUSIONS: Our study represents a comprehensive secretome analysis among parasitic plants and revealed both similarities and differences in candidate VFs between Striga and Cuscuta species. This knowledge is crucial for the development of new management strategies and delaying the evolution of virulence in parasitic weeds.


Sujet(s)
Cuscuta , Parasites , Striga , Animaux , Striga/génétique , Cuscuta/génétique , Sécrétome , Facteurs de virulence/génétique , Mauvaises herbes
16.
Sci Rep ; 14(1): 8379, 2024 04 10.
Article de Anglais | MEDLINE | ID: mdl-38600116

RÉSUMÉ

Macrophage responses to activation are fluid and dynamic in their ability to respond appropriately to challenges, a role integral to host defence. While bacteria can influence macrophage differentiation and polarization into pro-inflammatory and alternatively activated phenotypes through direct interactions, many questions surround indirect communication mechanisms mediated through secretomes derived from gut bacteria, such as lactobacilli. We examined effects of secretome-mediated conditioning on THP-1 human monocytes, focusing on the ability of the Lacticaseibacillus rhamnosus R0011 secretome (LrS) to drive macrophage differentiation and polarization and prime immune responses to subsequent challenge with lipopolysaccharide (LPS). Genome-wide transcriptional profiling revealed increased M2-associated gene transcription in response to LrS conditioning in THP-1 cells. Cytokine and chemokine profiling confirmed these results, indicating increased M2-associated chemokine and cytokine production (IL-1Ra, IL-10). These cells had increased cell-surface marker expression of CD11b, CD86, and CX3CR1, coupled with reduced expression of the M1 macrophage-associated marker CD64. Mitochondrial substrate utilization assays indicated diminished reliance on glycolytic substrates, coupled with increased utilization of citric acid cycle intermediates, characteristics of functional M2 activity. LPS challenge of LrS-conditioned THP-1s revealed heightened responsiveness, indicative of innate immune priming. Resting stage THP-1 macrophages co-conditioned with LrS and retinoic acid also displayed an immunoregulatory phenotype with expression of CD83, CD11c and CD103 and production of regulatory cytokines. Secretome-mediated conditioning of macrophages into an immunoregulatory phenotype is an uncharacterized and potentially important route through which lactic acid bacteria and the gut microbiota may train and shape innate immunity at the gut-mucosal interface.


Sujet(s)
Lacticaseibacillus rhamnosus , Monocytes , Humains , Monocytes/métabolisme , Sécrétome , Lipopolysaccharides , Cytokines/métabolisme , Chimiokines/métabolisme , Immunité
17.
Cells ; 13(7)2024 Mar 25.
Article de Anglais | MEDLINE | ID: mdl-38607010

RÉSUMÉ

Extracellular vesicles (EVs) are secreted from many tumors, including glioblastoma multiforme (GBM), the most common and lethal brain tumor in adults, which shows high resistance to current therapies and poor patient prognosis. Given the high relevance of the information provided by cancer cell secretome, we performed a proteomic analysis of microvesicles (MVs) and exosomes (EXOs) released from GBM-derived stem cells (GSCs). The latter, obtained from the brain of GBM patients, expressed P2X7 receptors (P2X7Rs), which positively correlate with GBM growth and invasiveness. P2X7R stimulation of GSCs caused significant changes in the EV content, mostly ex novo inducing or upregulating the expression of proteins related to cytoskeleton reorganization, cell motility/spreading, energy supply, protection against oxidative stress, chromatin remodeling, and transcriptional regulation. Most of the induced/upregulated proteins have already been identified as GBM diagnostic/prognostic factors, while others have only been reported in peripheral tumors. Our findings indicate that P2X7R stimulation enhances the transport and, therefore, possible intercellular exchange of GBM aggressiveness-increasing proteins by GSC-derived EVs. Thus, P2X7Rs could be considered a new druggable target of human GBM, although these data need to be confirmed in larger experimental sets.


Sujet(s)
Vésicules extracellulaires , Glioblastome , Récepteurs purinergiques P2X7 , Sécrétome , Humains , Lignée cellulaire tumorale , Vésicules extracellulaires/métabolisme , Glioblastome/métabolisme , Cellules souches tumorales/anatomopathologie , Protéome/métabolisme , Protéomique , Récepteurs purinergiques P2X7/métabolisme
18.
Int J Mol Sci ; 25(8)2024 Apr 14.
Article de Anglais | MEDLINE | ID: mdl-38673927

RÉSUMÉ

Domestic cat blastocysts cultured without the zona pellucida exhibit reduced implantation capacity. However, the protein expression profile has not been evaluated in these embryos. The objective of this study was to evaluate the protein expression profile of domestic cat blastocysts cultured without the zona pellucida. Two experimental groups were generated: (1) domestic cat embryos generated by IVF and cultured in vitro (zona intact, (ZI)) and (2) domestic cat embryos cultured in vitro without the zona pellucida (zona-free (ZF group)). The cleavage, morula, and blastocyst rates were estimated at days 2, 5 and 7, respectively. Day 7 blastocysts and their culture media were subjected to liquid chromatography-tandem mass spectrometry (LC-MS/MS). The UniProt Felis catus database was used to identify the standard proteome. No significant differences were found in the cleavage, morula, or blastocyst rates between the ZI and ZF groups (p > 0.05). Proteomic analysis revealed 22 upregulated and 20 downregulated proteins in the ZF blastocysts. Furthermore, 14 proteins involved in embryo development and implantation were present exclusively in the culture medium of the ZI blastocysts. In conclusion, embryo culture without the zona pellucida did not affect in vitro development, but altered the protein expression profile and release of domestic cat blastocysts.


Sujet(s)
Blastocyste , Protéomique , Zone pellucide , Animaux , Blastocyste/métabolisme , Zone pellucide/métabolisme , Chats , Protéomique/méthodes , Techniques de culture d'embryons , Sécrétome/métabolisme , Femelle , Fécondation in vitro , Protéome/métabolisme , Développement embryonnaire , Spectrométrie de masse en tandem , Chromatographie en phase liquide
19.
Stem Cell Res Ther ; 15(1): 121, 2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38664697

RÉSUMÉ

BACKGROUND: Triple-negative breast cancer (TNBC) is the most lethal subtype of breast cancer and, despite its adverse effects, chemotherapy is the standard systemic treatment option for TNBC. Since, it is of utmost importance to consider the combination of different agents to achieve greater efficacy and curability potential, MSC secretome is a possible innovative alternative. METHODS: In the present study, we proposed to investigate the anti-tumor effect of the combination of a chemical agent (paclitaxel) with a complex biological product, secretome derived from human Uterine Cervical Stem cells (CM-hUCESC) in TNBC. RESULTS: The combination of paclitaxel and CM-hUCESC decreased cell proliferation and invasiveness of tumor cells and induced apoptosis in vitro (MDA-MB-231 and/or primary tumor cells). The anti-tumor effect was confirmed in a mouse tumor xenograft model showing that the combination of both products has a significant effect in reducing tumor growth. Also, pre-conditioning hUCESC with a sub-lethal dose of paclitaxel enhances the effect of its secretome and in combination with paclitaxel reduced significantly tumor growth and even allows to diminish the dose of paclitaxel in vivo. This effect is in part due to the action of extracellular vesicles (EVs) derived from CM-hUCESC and soluble factors, such as TIMP-1 and - 2. CONCLUSIONS: In conclusion, our data demonstrate the synergistic effect of the combination of CM-hUCESC with paclitaxel on TNBC and opens an opportunity to reduce the dose of the chemotherapeutic agents, which may decrease chemotherapy-related toxicity.


Sujet(s)
Prolifération cellulaire , Cellules souches mésenchymateuses , Paclitaxel , Sécrétome , Tumeurs du sein triple-négatives , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Humains , Femelle , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Sécrétome/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , Apoptose/effets des médicaments et des substances chimiques , Col de l'utérus/métabolisme , Col de l'utérus/anatomopathologie , Col de l'utérus/effets des médicaments et des substances chimiques
20.
Biomed Pharmacother ; 174: 116536, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38569274

RÉSUMÉ

Diabetic kidney disease (DKD) is a leading cause of kidney failure. However, the involvement of renal fibroblasts and their communications with renal epithelial cells during DKD remain poorly understood. We investigated the potential role of renal proximal tubular epithelial cells (PTECs) in renal fibroblast activation that might lead to DKD. Additionally, the protective effects of curcumin, a known antioxidant, against renal fibroblast activation induced by high glucose-treated PTECs were investigated. Secretome was collected from HK-2 PTECs under normal glucose, high glucose, high glucose pretreated/cotreated with curcumin, or osmotic control condition for 24 h. Such secretome was then used to treat BHK-21 renal fibroblasts for 24 h. BHK-21 cells treated with high glucose-induced secretome had increased levels of fibroblast activation markers, including spindle index, F-actin, α-smooth muscle actin (α-SMA), fibronectin, collagen I, matrix metalloproteinase-2 (MMP-2) and MMP-9, as compared with normal glucose and osmotic control conditions. However, all these increases were successfully mitigated by curcumin. In addition, high glucose markedly increased intracellular reactive oxygen species (ROS) and transforming growth factor-ß (TGF-ß) secretion, but did not affect the secretion of platelet-derived growth factor A (PDGFA) and interleukin-1ß (IL-1ß), in HK-2 renal cells as compared with normal glucose and osmotic control conditions. Both intracellular ROS and secreted TGF-ß levels were successfully mitigated by curcumin. Therefore, curcumin prevents the high glucose-induced stimulatory effects of renal cell secretome on fibroblast activation, at least in part, via mitigating intracellular ROS and TGF-ß secretion.


Sujet(s)
Curcumine , Fibroblastes , Glucose , Espèces réactives de l'oxygène , Facteur de croissance transformant bêta , Curcumine/pharmacologie , Glucose/toxicité , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Facteur de croissance transformant bêta/métabolisme , Humains , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Animaux , Sécrétome/effets des médicaments et des substances chimiques , Sécrétome/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Rein/anatomopathologie , Néphropathies diabétiques/métabolisme , Antioxydants/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...