Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Front Immunol ; 15: 1367432, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38994364

RESUMEN

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.


Asunto(s)
Células Epiteliales , Neutrófilos , Secretoma , Animales , Bovinos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/inmunología , Secretoma/metabolismo , Femenino , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Fagocitosis , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/citología , Células Cultivadas , Especies Reactivas de Oxígeno/metabolismo
2.
Theriogenology ; 224: 107-118, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38761667

RESUMEN

Mesenchymal stromal cells (MSCs) have regenerative and immunomodulatory potential and may be used to treat injured tissues. Pregnancy has been associated with increased MSCs in the peripheral circulation in multiple species, but to date, there are no reports on this matter in horses. This study aimed to evaluate the effect of pregnancy on isolation efficiency and proliferation capacity of equine MSCs derived from the peripheral blood (PB) of mares. Venous blood samples were collected at the 11th month of gestation and 1 month after delivery from clinically healthy Arabian mares that presented normal pregnancies. Blood samples were processed for in vitro cellular culture and hormonal and metabolic profiles. MSCs were isolated and characterized by trilineage differentiation potential, immunophenotyping, analyzed by gene sequencing and proliferation assays. The isolation of peripheral blood mononuclear cells (PBMCs) of pregnant mares were associated with higher isolation efficiency and proliferative capacity of MSCs derived from peripheral blood (PB-MSCs) recovered pre-partum than those isolated post-partum. Although fetal gender, parity, 5α-reduced pregnanes, insulin, and cortisol were shown to affect cellular proliferation, individual factors and the small population studied must be considered. This study suggests that PB-MSCs from pregnant mares could be a valuable alternative source of MSCs for therapeutic purposes.


Asunto(s)
Proliferación Celular , Células Madre Mesenquimatosas , Animales , Femenino , Caballos , Embarazo , Células Madre Mesenquimatosas/fisiología , Células Madre Mesenquimatosas/citología , Preñez , Leucocitos Mononucleares/fisiología , Diferenciación Celular , Células Cultivadas
3.
Chembiochem ; 25(9): e202300822, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38487927

RESUMEN

This review focuses on discussing natural products (NPs) that contain higher homologated amino acids (homoAAs) in the structure as well as the proposed and characterized biosynthesis of these non-proteinogenic amino acids. Homologation of amino acids includes the insertion of a methylene group into its side chain. It is not a very common modification found in NP biosynthesis as approximately 450 homoAA-containing NPs have been isolated from four bacterial phyla (Cyanobacteria, Actinomycetota, Myxococcota, and Pseudomonadota), two fungal phyla (Ascomycota and Basidiomycota), and one animal phylum (Porifera), except for a few examples. Amino acids that are found to be homologated and incorporated in the NP structures include the following ten amino acids: alanine, arginine, cysteine, isoleucine, glutamic acid, leucine, phenylalanine, proline, serine, and tyrosine, where isoleucine, leucine, phenylalanine, and tyrosine share the comparable enzymatic pathway. Other amino acids have their individual homologation pathway (arginine, proline, and glutamic acid for bacteria), likely utilize the primary metabolic pathway (alanine and glutamic acid for fungi), or have not been reported (cysteine and serine). Despite its possible high potential in the drug discovery field, the biosynthesis of homologated amino acids has a large room to explore for future combinatorial biosynthesis and metabolic engineering purpose.


Asunto(s)
Aminoácidos , Productos Biológicos , Productos Biológicos/química , Productos Biológicos/metabolismo , Aminoácidos/química , Aminoácidos/metabolismo , Bacterias/metabolismo , Hongos/metabolismo , Hongos/química , Animales , Poríferos
4.
PLoS One ; 19(2): e0298390, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38416747

RESUMEN

Although highly conserved in structure and function, many (patho)physiological processes of the mammary gland vary drastically between mammals, with mechanisms regulating these differences not well understood. Large mammals display variable lactation strategies and mammary cancer incidence, however, research into these variations is often limited to in vitro analysis due to logistical limitations. Validating a model with functional mammary xenografts from cryopreserved tissue fragments would allow for in vivo comparative analysis of mammary glands from large and/or rare mammals and would improve our understanding of postnatal development, lactation, and premalignancy across mammals. To this end, we generated functional mammary xenografts using mammary tissue fragments containing mammary stroma and parenchyma isolated via an antibody-independent approach from healthy, nulliparous equine and canine donor tissues to study these species in vivo. Cryopreserved mammary tissue fragments were xenotransplanted into de-epithelialized fat pads of immunodeficient mice and resulting xenografts were structurally and functionally assessed. Preimplantation of mammary stromal fibroblasts was performed to promote ductal morphogenesis. Xenografts recapitulated mammary lobule architecture and contained donor-derived stromal components. Mammatropic hormone stimulation resulted in (i) upregulation of lactation-associated genes, (ii) altered proliferation index, and (iii) morphological changes, indicating functionality. Preimplantation of mammary stromal fibroblasts did not promote ductal morphogenesis. This model presents the opportunity to study novel mechanisms regulating unique lactation strategies and mammary cancer induction in vivo. Due to the universal applicability of this approach, this model serves as proof-of-concept for developing mammary xenografts for in vivo analysis of virtually any mammals, including large and rare mammals.


Asunto(s)
Neoplasias de la Mama , Glándulas Mamarias Humanas , Humanos , Femenino , Ratones , Animales , Caballos , Perros , Trasplante Heterólogo , Glándulas Mamarias Animales/patología , Lactancia/fisiología , Mamíferos , Neoplasias de la Mama/patología
5.
Commun Biol ; 6(1): 1006, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37789172

RESUMEN

Mammary cancer incidence varies greatly across species and underlying mechanisms remain elusive. We previously showed that mammosphere-derived epithelial cells from species with low mammary cancer incidence, such as horses, respond to carcinogen 7, 12-Dimethylbenz(a)anthracene-induced DNA damage by undergoing apoptosis, a postulated anti-cancer mechanism. Additionally, we found that miR-214-3p expression in mammosphere-derived epithelial cells is lower in mammary cancer-resistant as compared to mammary cancer-susceptible species. Here we show that increasing miR-214 expression and decreasing expression of its target gene nuclear factor kappa B subunit 1 in mammosphere-derived epithelial cells from horses abolishes 7,12-Dimethylbenz(a)anthracene-induced apoptosis. A direct interaction of miR-214-3p with another target gene, unc-5 netrin receptor A, is also demonstrated. We propose that relatively low levels of miR-214 in mammosphere-derived epithelial cells from mammals with low mammary cancer incidence, allow for constitutive gene nuclear factor kappa B subunit 1 expression and apoptosis in response to 7, 12-Dimethylbenz(a)anthracene. Better understanding of the mechanisms regulating cellular responses to carcinogens improves our overall understanding of mammary cancer resistance mechanisms.


Asunto(s)
MicroARNs , Neoplasias , Animales , Caballos , Carcinógenos/toxicidad , Carcinógenos/metabolismo , 9,10-Dimetil-1,2-benzantraceno/toxicidad , 9,10-Dimetil-1,2-benzantraceno/metabolismo , FN-kappa B/metabolismo , Células Epiteliales/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Apoptosis , Antracenos/metabolismo , Antracenos/farmacología , Mamíferos , Neoplasias/metabolismo
6.
Sci Rep ; 13(1): 12600, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37537239

RESUMEN

The bovine mammary stem/progenitor cell secretome stimulates regeneration in vitro and contains proteins associated with antimicrobial defense. This has led to the exploration of the secretome as a biologic treatment for mastitis, a costly inflammation of the udder commonly caused by bacteria. This study reports on a population of bovine mammary stem/progenitor cells isolated non-invasively from milk (MiDCs). MiDCs were characterized by immunophenotyping, mammosphere formation assays, and single cell RNA sequencing. They displayed epithelial morphology, exhibited markers of mammary stem/progenitor cells, and formed mammospheres, like mammary gland tissue-isolated stem/progenitor cells. Single cell RNA sequencing revealed two sub-populations of MiDCs: epithelial cells and macrophages. Functionally, the MiDC secretome increased fibroblast migration, promoted angiogenesis of endothelial cells, and inhibited the growth of mastitis-associated bacteria, including antibiotic-resistant strains, in vitro. These qualities of MiDCs render them a source of stem cells and stem cell products that may be used to treat diseases affecting the dairy industry, including mastitis.


Asunto(s)
Mastitis , Leche , Femenino , Animales , Humanos , Leche/metabolismo , Transcriptoma , Células Endoteliales , Células Epiteliales/metabolismo , Bacterias , Glándulas Mamarias Animales/metabolismo
7.
J Mammary Gland Biol Neoplasia ; 27(2): 185-210, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35904679

RESUMEN

Mammary cancer, or breast cancer in women, is a polygenic disease with a complex etiopathogenesis. While much remains elusive regarding its origin, it is well established that chemical carcinogens and endogenous estrogens contribute significantly to the initiation and progression of this disease. Rats have been useful models to study induced mammary cancer. They develop mammary tumors with comparable histopathology to humans and exhibit differences in resistance or susceptibility to mammary cancer depending on strain. While some rat strains (e.g., Sprague-Dawley) readily form mammary tumors following treatment with the chemical carcinogen, 7,12-dimethylbenz[a]-anthracene (DMBA), other strains (e.g., Copenhagen) are resistant to DMBA-induced mammary carcinogenesis. Genetic linkage in inbred strains has identified strain-specific quantitative trait loci (QTLs) affecting mammary tumors, via mechanisms that act together to promote or attenuate, and include 24 QTLs controlling the outcome of chemical induction, 10 QTLs controlling the outcome of estrogen induction, and 4 QTLs controlling the outcome of irradiation induction. Moreover, and based on shared factors affecting mammary cancer etiopathogenesis between rats and humans, including orthologous risk regions between both species, rats have served as useful models for identifying methods for breast cancer prediction and treatment. These studies in rats, combined with alternative animal models that more closely mimic advanced stages of breast cancer and/or human lifestyles, will further improve our understanding of this complex disease.


Asunto(s)
Neoplasias de la Mama , Neoplasias Mamarias Animales , Neoplasias Mamarias Experimentales , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Neoplasias de la Mama/genética , Carcinógenos , Estrógenos/genética , Femenino , Humanos , Neoplasias Mamarias Animales/inducido químicamente , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Sitios de Carácter Cuantitativo , Ratas , Ratas Sprague-Dawley
8.
Development ; 149(7)2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35297994

RESUMEN

Mammary organoid (MaO) models are only available for a few traditional model organisms, limiting our ability to investigate mammary gland development and cancer across mammals. This study established equine mammary organoids (EqMaOs) from cryopreserved mammary tissue, in which mammary tissue fragments were isolated and embedded into a 3D matrix to produce EqMaOs. We evaluated viability, proliferation and budding capacity of EqMaOs at different time points during culture, showing that although the number of proliferative cells decreased over time, viability was maintained and budding increased. We further characterized EqMaOs based on expression of stem cell, myoepithelial and luminal markers, and found that EqMaOs expressed these markers throughout culture and that a bilayered structure as seen in vivo was recapitulated. We used the milk-stimulating hormone prolactin to induce milk production, which was verified by the upregulation of milk proteins, most notably ß-casein. Additionally, we showed that our method is also applicable to additional non-traditional mammalian species, particularly domesticated animals such as cats, pigs and rabbits. Collectively, MaO models across species will be a useful tool for comparative developmental and cancer studies.


Asunto(s)
Glándulas Mamarias Animales , Organoides , Animales , División Celular , Células Epiteliales/metabolismo , Femenino , Caballos , Lactancia , Mamíferos , Conejos , Células Madre , Porcinos
9.
Res Vet Sci ; 141: 76-80, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34700147

RESUMEN

Equid herpesvirus 1 (EHV-1) outbreaks occur when virus spreads from infected horses to in-contact horses, primarily via nasal shedding. This study evaluated the efficacy of factors secreted by equine peripheral blood derived mesenchymal stromal cells (PB-MSCs), collectively named the secretome, to inhibit the growth of EHV-1 in (i) 2D epithelial cell cultures (RK-13) in vitro, (ii) 3D equine nasal explants in vitro and (iii) an EHV-1 infection mouse model in vivo. The PB-MSC secretome was found to inhibit EHV-1 in RK-13 cells as well as in the epithelium of equine nasal explants. Although the PB-MSC secretome did not decrease overall severity of EHV-1 infection in mice, as determined by weight loss and viral titers in lungs, histological analyses indicated local reduction of EHV-1 infection in nasal epithelium. These results indicate that the PB-MSC secretome inhibits EHV-1 in epithelial cells in a context-dependent manner.


Asunto(s)
Infecciones por Herpesviridae , Herpesvirus Équido 1 , Herpesvirus Équido 4 , Enfermedades de los Caballos , Células Madre Mesenquimatosas , Enfermedades de los Roedores , Animales , Células Epiteliales , Infecciones por Herpesviridae/veterinaria , Caballos , Ratones , Mucosa Nasal , Secretoma
10.
Stem Cells Transl Med ; 10(12): 1666-1679, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34528765

RESUMEN

Mesenchymal stromal cells (MSCs) from both humans and horses, which represent a clinically relevant translation animal model for human cutaneous wound healing, were recently found to possess antimicrobial properties against planktonic bacteria, and in the case of equine MSCs, also against biofilms. This, together with previous findings that human and equine MSCs promote angiogenesis and wound healing, makes these cells an attractive approach to treat infected cutaneous wounds in both species. The anti-biofilm activities of equine MSC, via secretion of cysteine proteases, have only been demonstrated in vitro, thus lacking information about in vivo relevance. Moreover, the effects of the equine MSC secretome on resident skin cells have not yet been explored. The goals of this study were to (a) test the efficacy of the MSC secretome in a physiologically relevant ex vivo equine skin biofilm explant model and (b) explore the impact of the MSC secretome on the antimicrobial defense mechanisms of resident skin cells. Our salient findings were that secreted factors from equine MSCs significantly decreased viability of methicillin-resistant Staphylococcus aureus bacteria in mature biofilms in this novel skin biofilm explant model. Moreover, we demonstrated that equine MSCs secrete CCL2 that increases the antimicrobial activity of equine keratinocytes by stimulating expression of antimicrobial peptides. Collectively, these data contribute to our understanding of the MSC secretome's antimicrobial properties, both directly by killing bacteria and indirectly by stimulating immune responses of surrounding resident skin cells, thus further supporting the value of MSC secretome-based treatments for infected wounds.


Asunto(s)
Células Madre Mesenquimatosas , Staphylococcus aureus Resistente a Meticilina , Animales , Antibacterianos/metabolismo , Antibacterianos/farmacología , Péptidos Antimicrobianos , Mecanismos de Defensa , Modelos Animales de Enfermedad , Caballos , Queratinocitos , Células Madre Mesenquimatosas/metabolismo
11.
Cancer Metastasis Rev ; 40(1): 47-69, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33111160

RESUMEN

Traditional laboratory model organisms are indispensable for cancer research and have provided insight into numerous mechanisms that contribute to cancer development and progression in humans. However, these models do have some limitations, most notably related to successful drug translation, because traditional model organisms are often short-lived, small-bodied, genetically homogeneous, often immunocompromised, are not exposed to natural environments shared with humans, and usually do not develop cancer spontaneously. We propose that assimilating information from a variety of long-lived, large, genetically diverse, and immunocompetent species that live in natural environments and do develop cancer spontaneously (or do not develop cancer at all) will lead to a more comprehensive understanding of human cancers. These non-traditional model organisms can also serve as sentinels for environmental risk factors that contribute to human cancers. Ultimately, expanding the range of animal models that can be used to study cancer will lead to improved insights into cancer development, progression and metastasis, tumor microenvironment, as well as improved therapies and diagnostics, and will consequently reduce the negative impacts of the wide variety of cancers afflicting humans overall.


Asunto(s)
Neoplasias , Animales , Humanos , Modelos Animales , Neoplasias/etiología , Investigación , Microambiente Tumoral
12.
Stem Cell Res Ther ; 11(1): 524, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33276815

RESUMEN

BACKGROUND: The efficacy of mesenchymal stromal cell (MSC) therapy is thought to depend on the intrinsic heterogeneity of MSC cultures isolated from different tissue sources as well as individual MSCs isolated from the same tissue source, neither of which is well understood. To study this, we used MSC cultures isolated from horses. The horse is recognized as a physiologically relevant large animal model appropriate for translational MSC studies. Moreover, due to its large size the horse allows for the simultaneous collection of adequate samples from multiple tissues of the same animal, and thus, for the unique collection of donor matched MSC cultures from different sources. The latter is much more challenging in mice and humans due to body size and ethical constraints, respectively. METHODS: In the present study, we performed single-cell RNA sequencing (scRNA-seq) on primary equine MSCs that were collected from three donor-matched tissue sources; adipose tissue (AT), bone marrow (BM), and peripheral blood (PB). Based on transcriptional differences detected with scRNA-seq, we performed functional experiments to examine motility and immune regulatory function in distinct MSC populations. RESULTS: We observed both inter- and intra-source heterogeneity across the three sources of equine MSCs. Functional experiments demonstrated that transcriptional differences correspond with phenotypic variance in cellular motility and immune regulatory function. Specifically, we found that (i) differential expression of junctional adhesion molecule 2 (JAM2) between MSC cultures from the three donor-matched tissue sources translated into altered cell motility of BM-derived MSCs when RNA interference was used to knock down this gene, and (ii) differences in C-X-C motif chemokine ligand 6 (CXCL6) expression in clonal MSC lines derived from the same tissue source correlated with the chemoattractive capacity of PB-derived MSCs. CONCLUSIONS: Ultimately, these findings will enhance our understanding of MSC heterogeneity and will lead to improvements in the therapeutic potential of MSCs, accelerating the transition from bench to bedside.


Asunto(s)
Células Madre Mesenquimatosas , Animales , Células de la Médula Ósea , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Caballos , Ratones , Análisis de Secuencia de ARN
13.
Sci Rep ; 10(1): 20580, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33239740

RESUMEN

Determining mechanisms that naturally protect species from developing cancer is critical in order to prevent and treat cancer. Here, we describe a novel cancer-suppressing mechanism, via the secretion of bioactive factors by mammary cells, that is present in domesticated mammals with a low mammary cancer incidence. Specifically, these bioactive factors induced triple-negative breast cancer cell (TNBC) death in vitro and reduced tumorigenicity in a xenograft TNBC mouse model in vivo. RNA deep sequencing showed significant downregulation of genes associated with breast cancer progression in secretome-cultured TNBC cells. Further in-depth multi-omics analysis identified sphingomyelins as key secreted factors, and their role was confirmed via inhibition of the sphingomyelin signaling pathway. We speculate that secreted sphingomyelins in the mammary gland of mammals with a naturally low incidence of mammary cancer mediate the elimination of cancer cells. This study contributes to the growing list of protective mechanisms identified in cancer-proof species.


Asunto(s)
Neoplasias de la Mama/metabolismo , Esfingomielinas/metabolismo , Esfingomielinas/farmacología , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Caballos , Humanos , Incidencia , Ratones , Ratones Desnudos , Transducción de Señal/genética , Esfingomielinas/fisiología , Neoplasias de la Mama Triple Negativas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Stem Cells Transl Med ; 9(7): 746-757, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32216094

RESUMEN

Mesenchymal stromal cells (MSCs) from various species, such as humans, mice, and horses, were recently found to effectively inhibit the growth of various bacteria associated with chronic infections, such as nonhealing cutaneous wounds, via secretion of antimicrobial peptides. These MSC antimicrobial properties have primarily been studied in the context of the planktonic phenotype, and thus, information on the effects on bacteria in biofilms is largely lacking. The objectives of this study were to evaluate the in vitro efficacy of the MSC secretome against various biofilm-forming wound pathogens, including the methicillin-resistant Staphylococcus aureus (MRSA), and to explore the mechanisms that affect bacterial biofilms. To this end, we used equine MSCs, because the horse represents a physiologically relevant model for human wound healing and offers a readily translatable model for MSC therapies in humans. Our salient findings were that the equine MSC secretome inhibits biofilm formation and mature biofilms of various bacteria, such as Pseudomonas aeruginosa, S. aureus, and Staphylococcus epidermidis. Furthermore, we demonstrated that equine MSC secrete cysteine proteases that destabilize MRSA biofilms, thereby increasing the efficacy of antibiotics that were previously tolerated by the biofilms. In light of the rise of antibiotic-resistant bacterial strains as an increasing global health threat, our results provide the rationale for using the MSC secretome as a complementary treatment for bacterial skin infections in both humans and horses.


Asunto(s)
Biopelículas/efectos de los fármacos , Proteasas de Cisteína/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Caballos
15.
Vet Immunol Immunopathol ; 202: 25-30, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30078595

RESUMEN

Mesenchymal stromal cells (MSC) have the therapeutic potential to decrease inflammation due to their immunomodulatory properties. They can be isolated from various tissue sources such as bone marrow, adipose tissue, and blood, but it is unknown how the tissue source of origin affects the responses of MSC to inflammatory stimuli. Here, we conceptually addressed this question by evaluating the immune-related gene expression profiles of equine MSC from different tissue sources in response to interferon gamma (IFN-γ) stimulation, with the goal to determine if there is a preferable MSC source for clinical application in an inflammatory environment. The salient findings from this initial study were that the baseline expression of all immune related genes analyzed, with the exception of prostaglandin-endoperoxide synthase 2 (PTGS2), was variable in MSC depending on tissue source. Following IFN-γ stimulation, however, gene expression profiles became more similar across all tissue sources, suggesting that MSC from different sources will likely respond similarly in an inflammatory environment when used clinically.


Asunto(s)
Inflamación/veterinaria , Interferón gamma/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Animales , Diferenciación Celular/genética , Proliferación Celular , Células Cultivadas , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/inmunología , Caballos/inmunología , Inmunomodulación , Transcriptoma
16.
Cytotherapy ; 20(8): 1061-1076, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30087008

RESUMEN

BACKGROUND: Impaired cutaneous wound healing is common in humans, and treatments are often ineffective. Based on the significant emotional and economic burden of impaired wound healing, innovative therapies are needed. The potential of mesenchymal stromal cell (MSC)-secreted factors to treat cutaneous wounds is an active area of research that is in need of refinement before effective clinical trials can be initiated. The aims of the present study were to (i) study which MSC-secreted factors stimulate dermal fibroblast (DF) migration in vitro and (ii) evaluate the potential of these factors to promote wound healing in vivo. METHODS: To this end, MSCs were isolated from the peripheral blood of healthy horses, a physiologically relevant large animal model appropriate for translational wound-healing studies. Conditioned medium (CM) from cultured equine MSCs was analyzed using liquid chromatography-mass spectrophotometry (LC-MS/MS) to identify secreted proteins of interest. Double-stranded RNA-mediated interference (RNAi) was used to silence the genes encoding selected proteins, and the effects of CM from these transfected MSCs on migration of cultured equine DF cells in vitro and full-thickness wounds in mice were evaluated. RESULTS: We found that MSC-derived plasminogen activator inhibitor-1 (PAI-1) and tenascin-C significantly increased DF migration in vitro and improved wound healing in vivo by decreasing time to wound closure. DISCUSSION: These results suggest that in a complex wound environment, MSC-secreted factors PAI-1 and tenascin-C contribute to the positive effect of therapeutically applied MSC CM on wound healing.


Asunto(s)
Dermis/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Inhibidor 1 de Activador Plasminogénico , Tenascina , Cicatrización de Heridas/efectos de los fármacos , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Dermis/citología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/fisiología , Femenino , Fibroblastos/fisiología , Caballos , Células Madre Mesenquimatosas/citología , Ratones , Células 3T3 NIH , Inhibidor 1 de Activador Plasminogénico/metabolismo , Inhibidor 1 de Activador Plasminogénico/farmacología , Tenascina/metabolismo , Tenascina/farmacología , Cicatrización de Heridas/fisiología
17.
Neurotoxicology ; 67: 54-64, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29660375

RESUMEN

Arsenic exposure through contaminated food, water, and air causes irreversible neural damage and affects millions of people worldwide. Several studies have demonstrated that the secreted factors (secretome) from mesenchymal stromal/stem cells (MSCs) can promote neural recovery after several forms of injury including stroke and neurodegenerative diseases. The present study was conducted to determine if the secretome from adipose-derived MSCs (ADSCs) prevents arsenic damage to SH-SY5Y cells. To this end, human neuroblastoma cells (SH-SY5Y) were pre-treated with the secretome from ADSCs and then challenged with different concentrations of arsenic. After various doses and exposure times, the extent of neuronal injury was assessed using MTT reduction and LDH release assays as well as LIVE/DEAD staining. These data demonstrate that the ADSC secretome protects SH-SY5Y cells from arsenic-induced toxicity. Previous reports have shown that the secretome of MSCs can induce neuroblast differentiation and mature neurons are less susceptible to chemical-induced toxicity. In the current study, proliferation assays, neurite length assessment, and quantitative RT-PCR of differentiation markers indicated that the ADSC secretome does not induce SH-SY5Y differentiation into a mature neuron-like phenotype. In contrast, our results demonstrated that soluble factor(s) in the ADSC secretome enhance SH-SY5Y cell substrate-dependent adhesion. The present study is the first to illustrate that the secretome from ADSCs protects SH-SY5Y cells from arsenic-induced toxicity. Additionally, we showed that protection against arsenic toxicity is not dependent on SH-SY5Y cell differentiation into a mature neuron-like phenotype, but involves soluble factor(s) in the secretome that appear to enhance cell survival by an adhesion-dependent mechanism.


Asunto(s)
Tejido Adiposo/metabolismo , Arsénico/toxicidad , Diferenciación Celular/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Diferenciación Celular/fisiología , Línea Celular Tumoral , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Humanos , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Neuronas/patología
18.
Stem Cell Res Ther ; 8(1): 157, 2017 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-28676123

RESUMEN

BACKGROUND: The prevalence of chronic skin wounds in humans is high, and treatment is often complicated by the presence of pathogenic bacteria. Therefore, safe and innovative treatments to reduce the bacterial load in cutaneous wounds are needed. Mesenchymal stromal cells (MSC) are known to provide paracrine signals that act on resident skin cells to promote wound healing, but their potential antibacterial activities are not well described. The present study was designed to examine the antibacterial properties of MSC from horses, as this animal model offers a readily translatable model for MSC therapies in humans. Specifically, we aimed to (i) evaluate the in vitro effects of equine MSC on the growth of representative gram-negative and gram-positive bacterial species commonly found in skin wounds and (ii) define the mechanisms by which MSC inhibit bacterial growth. METHODS: MSC were isolated from the peripheral blood of healthy horses. Gram-negative E. coli and gram-positive S. aureus were cultured in the presence of MSC and MSC conditioned medium (CM), containing all factors secreted by MSC. Bacterial growth was measured by plating bacteria and counting viable colonies or by reading the absorbance of bacterial cultures. Bacterial membrane damage was detected by incorporation of N-phenyl-1-naphthylamine (NPN). Antimicrobial peptide (AMP) gene and protein expression by equine MSC were determined by RT-PCR and Western blot analysis, respectively. Blocking of AMP activity of MSC CM was achieved using AMP-specific antibodies. RESULTS: We found that equine MSC and MSC CM inhibit the growth of E. coli and S. aureus, and that MSC CM depolarizes the cell membranes of these bacteria. In addition, we found that equine MSC CM contains AMPs, and blocking these AMPs with antibodies reduces the effects of MSC CM on bacteria. CONCLUSIONS: Our results demonstrate that equine MSC inhibit bacterial growth and secrete factors that compromise the membrane integrity of bacteria commonly found in skin wounds. We also identified four specific AMPs produced by equine MSC. The secretion of AMPs may contribute to the value of MSC as a therapy for cutaneous wounds in both horses and humans.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Infecciones por Escherichia coli/metabolismo , Escherichia coli/metabolismo , Células Madre Mesenquimatosas/metabolismo , Infecciones Cutáneas Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Infección de Heridas , Animales , Infecciones por Escherichia coli/patología , Caballos , Infecciones Cutáneas Estafilocócicas/patología , Infección de Heridas/metabolismo , Infección de Heridas/microbiología , Infección de Heridas/patología
19.
Wound Repair Regen ; 25(2): 234-247, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28370679

RESUMEN

The prevalence of cutaneous fibroproliferative disorders (CFPDs) is high and almost exclusively occurs in humans (keloids and hypertrophic scars) and horses (exuberant granulation tissue), making the horse a valuable translational model for studies on prevention and treatment of human CFPDs. CFPDs arise as a result of dysregulated wound healing characterized by persistently high levels of cytokines, such as transforming growth factor beta 1 (TGF-ß1), that contribute to excessive extracellular matrix deposition, and the physical disorganization of dermal fibroblasts (DF). The mesenchymal stromal cell (MSC) secretome, consisting of all factors secreted by MSC, has been shown to promote normal wound healing in both humans and horses, but its potential to treat CFPDs remains largely unexplored. Therefore, the objective of this study was to examine the effects of the equine MSC secretome on equine DF influenced by cytokines that contribute to the development of CFPDs. First, primary equine DF were treated with TGF-ß1 in vitro in the presence or absence of MSC secreted products. We found that MSC secreted products could block TGF-ß1-induced changes in DF morphology, proliferation rate, gene expression, and contractile-capacity. We then isolated primary DF from equine exuberant granulation tissue, to evaluate the potential of the MSC secretome to alter the phenotype of cells derived from a complex CFPD environment. These results showed that MSC secreted factors did not change proliferation or migration of these cells, but did lead to changes in expression of genes and proteins involved in extracellular matrix remodeling and did affect contractile capacity. These results warrant future studies designed to evaluate the potential of the MSC secretome to minimize the pathologies associated with CFPD in vivo.


Asunto(s)
Dermis/citología , Fibroblastos/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Heridas y Lesiones/patología , Heridas y Lesiones/terapia , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Tratamiento Basado en Trasplante de Células y Tejidos , Medios de Cultivo Condicionados , Fibroblastos/fisiología , Expresión Génica , Caballos , Células Madre Mesenquimatosas/citología , Modelos Animales , Investigación Biomédica Traslacional , Cicatrización de Heridas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA