Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 16.745
1.
BMC Cancer ; 24(1): 664, 2024 May 31.
Article En | MEDLINE | ID: mdl-38822331

Recent studies have shown that blue light-emitting diode (LED) light has anti-tumor effects, suggesting the possibility of using visible light in cancer therapy. However, the effects of blue light irradiation on cells in the tumor microenvironment, including tumor-associated macrophages (TAMs), are unknown. Here, THP-1 cells were cultured in the conditioned medium (CM) of HCT-116 cells to prepare TAMs. TAMs were divided into LED-irradiated and control groups. Then, the effects of blue LED irradiation on TAM activation were examined. Expression levels of M2 macrophage markers CD163 and CD206 expression were significantly decreased in LED-irradiated TAMs compared with the control group. While control TAM-CM could induce HCT-116 cell migration, these effects were not observed in cells cultured in TAM-CM with LED irradiation. Vascular endothelial growth factor (VEGF) secretion was significantly suppressed in LED-exposed TAMs. PD-L1 expression was upregulated in HCT-116 cells cultured with TAM-CM but attenuated in cells cultured with LED-irradiated TAM-CM. In an in vivo model, protein expression levels of F4/80 and CD163, which are TAM markers, were reduced in the LED-exposed group. These results indicate that blue LED light may have an inhibitory effect on TAMs, as well as anti-tumor effects on colon cancer cells.


Colonic Neoplasms , Light , Tumor-Associated Macrophages , Humans , Colonic Neoplasms/radiotherapy , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/radiation effects , Tumor-Associated Macrophages/immunology , Light/adverse effects , Animals , HCT116 Cells , Mice , Tumor Microenvironment/radiation effects , Cell Movement/radiation effects , Culture Media, Conditioned/pharmacology , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, CD/metabolism , Vascular Endothelial Growth Factor A/metabolism , Receptors, Cell Surface/metabolism , Macrophages/metabolism , Macrophages/radiation effects , Macrophages/immunology , Phototherapy/methods , Macrophage Activation/radiation effects , Blue Light
2.
Int J Med Sci ; 21(6): 1155-1164, 2024.
Article En | MEDLINE | ID: mdl-38774749

Introduction: Clinical studies have shown that endodontically-treated nonvital teeth exhibit less root resorption during orthodontic tooth movement. The purpose of this study was to explore whether hypoxic dental pulp stem cells (DPSCs) can promote osteoclastogenesis in orthodontically induced inflammatory root resorption (OIIRR). Methods: Succinate in the supernatant of DPSCs under normal and hypoxic conditions was measured by a succinic acid assay kit. The culture supernatant of hypoxia-treated DPSCs was used as conditioned medium (Hypo-CM). Bone marrow-derived macrophages (BMDMs) from succinate receptor 1 (SUCNR1)-knockout or wild-type mice were cultured with conditioned medium (CM), exogenous succinate or a specific inhibitor of SUCNR1 (4c). Tartrate-resistant acid phosphatase (TRAP) staining, Transwell assays, qPCR, Western blotting, and resorption assays were used to evaluate osteoclastogenesis-related changes. Results: The concentration of succinate reached a maximal concentration at 6 h in the supernatant of hypoxia-treated DPSCs. Hypo-CM-treated macrophages were polarized to M1 proinflammatory macrophages. Hypo-CM treatment significantly increased the formation and differentiation of osteoclasts and increased the expression of osteoclastogenesis-related genes, and this effect was inhibited by the specific succinate inhibitor 4c. Succinate promoted chemotaxis and polarization of M1-type macrophages with increased expression of osteoclast generation-related genes. SUCNR1 knockout decreased macrophage migration, M1 macrophage polarization, differentiation and maturation of osteoclasts, as shown by TRAP and NFATc1 expression and cementum resorption. Conclusions: Hypoxic DPSC-derived succinate may promote osteoclast differentiation and root resorption. The regulation of the succinate-SUCNR1 axis may contribute to the reduction in the OIIRR.


Dental Pulp , Mice, Knockout , Osteoclasts , Osteogenesis , Root Resorption , Stem Cells , Succinic Acid , Animals , Mice , Dental Pulp/cytology , Dental Pulp/drug effects , Dental Pulp/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Root Resorption/pathology , Root Resorption/metabolism , Humans , Succinic Acid/metabolism , Osteogenesis/drug effects , Stem Cells/metabolism , Stem Cells/drug effects , Cell Differentiation/drug effects , Macrophages/metabolism , Macrophages/drug effects , Cell Hypoxia/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Culture Media, Conditioned/pharmacology , Cells, Cultured
3.
Synapse ; 78(3): e22293, 2024 May.
Article En | MEDLINE | ID: mdl-38779935

The differentiation of bone marrow stromal cells (BMSCs) into Schwann-like cells (SCLCs) has the potential to promote the structural and functional restoration of injured axons. However, the optimal induction protocol and its underlying mechanisms remain unclear. This study aimed to compare the effectiveness of different induction protocols in promoting the differentiation of rat BMSCs into SCLCs and to explore their potential mechanisms. BMSCs were induced using two distinct methods: a composite factor induction approach (Protocol-1) and a conditioned culture medium induction approach (Protocol-2). The expression of Schwann cells (SCs) marker proteins and neurotrophic factors (NTFs) in the differentiated cells was assessed. Cell proliferation and apoptosis were also measured. During induction, changes in miR-21 and Sprouty RTK signaling antagonist 2 (SPRY2) mRNA were analyzed. Following the transfection of BMSCs with miR-21 agomir or miR-21 antagomir, induction was carried out using both protocols, and the expression of SPRY2, ERK1/2, and SCs marker proteins was examined. The results revealed that NTFs expression was higher in Protocol-1, whereas SCs marker proteins expression did not significantly differ between the two groups. Compared to Protocol-1, Protocol-2 exhibited enhanced cell proliferation and fewer apoptotic and necrotic cells. Both protocols showed a negative correlation between miR-21 and SPRY2 expression throughout the induction stages. After induction, the miR-21 agomir group exhibited reduced SPRY2 expression, increased ERK1/2 expression, and significantly elevated expression of SCs marker proteins. This study demonstrates that Protocol-1 yields higher NTFs expression, whereas Protocol-2 results in stronger SCLCs proliferation. Upregulating miR-21 suppresses SPRY2 expression, activates the ERK1/2 signaling pathway, and promotes BMSC differentiation into SCLCs.


Cell Differentiation , Cell Proliferation , Membrane Proteins , Mesenchymal Stem Cells , MicroRNAs , Rats, Sprague-Dawley , Schwann Cells , Animals , Schwann Cells/metabolism , Schwann Cells/cytology , MicroRNAs/metabolism , MicroRNAs/genetics , Cell Differentiation/physiology , Rats , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Proliferation/physiology , Cells, Cultured , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Apoptosis/physiology , Nerve Growth Factors/metabolism , Nerve Growth Factors/genetics , Culture Media, Conditioned/pharmacology , Nerve Tissue Proteins
4.
Neoplasma ; 71(2): 164-179, 2024 Apr.
Article En | MEDLINE | ID: mdl-38766857

Obesity is a major public health concern because it increases the risk of several diseases, including cancer. Crosstalk between obesity and cancer seems to be very complex, and the interaction between adipocytes and cancer cells leads to changes in adipocytes' function and their paracrine signaling, promoting a microenvironment that supports tumor growth. Carbonic anhydrase IX (CA IX) is a tumor-associated enzyme that not only participates in pH regulation but also facilitates metabolic reprogramming and supports the migration, invasion, and metastasis of cancer cells. In addition, CA IX expression, predominantly regulated via hypoxia-inducible factor (HIF-1), serves as a surrogate marker of hypoxia. In this study, we investigated the impact of adipocytes and adipocyte-derived factors on the expression of CA IX in colon and breast cancer cells. We observed increased expression of CA9 mRNA as well as CA IX protein in the presence of adipocytes and adipocyte-derived conditioned medium. Moreover, we confirmed that adipocytes affect the hypoxia signaling pathway and that the increased CA IX expression results from adipocyte-mediated induction of HIF-1α. Furthermore, we demonstrated that adipocyte-mediated upregulation of CA IX leads to increased migration and decreased adhesion of colon cancer cells. Finally, we brought experimental evidence that adipocytes, and more specifically leptin, upregulate CA IX expression in cancer cells and consequently promote tumor progression.


Adipocytes , Antigens, Neoplasm , Breast Neoplasms , Carbonic Anhydrase IX , Cell Movement , Colonic Neoplasms , Hypoxia-Inducible Factor 1, alpha Subunit , Leptin , Paracrine Communication , Humans , Carbonic Anhydrase IX/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Antigens, Neoplasm/metabolism , Female , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Leptin/metabolism , Cell Line, Tumor , Animals , Obesity/metabolism , Culture Media, Conditioned/pharmacology , Tumor Microenvironment , Gene Expression Regulation, Neoplastic , Mice
5.
Mol Biol Rep ; 51(1): 674, 2024 May 24.
Article En | MEDLINE | ID: mdl-38787497

BACKGROUND: Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) with inflammation and immune dysfunction. OBJECTIVES: We compared the remyelination and immunomodulation properties of mesenchymal stem cells (MSCs) with their conditioned medium (CM) in the cuprizone model. METHODS: Twenty-four C57BL/ 6 mice were divided into four groups. After cuprizone demyelination, MSCs and their CM were injected into the right lateral ventricle of mice. The expression level of IL-1ß, TNF-α, and BDNF genes was evaluated using the qRT-PCR. APC antibody was used to assess the oligodendrocyte population using the immunofluorescent method. The remyelination and axonal repair were studied by specific staining of the LFB and electron microscopy techniques. RESULTS: Transplantation of MSCs and CM increased the expression of the BDNF gene and decreased the expression of IL-1ß and TNF-α genes compared to the cuprizone group, and these effects in the cell group were more than CM. Furthermore, cell transplantation resulted in a significant improvement in myelination and axonal repair, which was measured by luxol fast blue and transmission electron microscope images. The cell group had a higher number of oligodendrocytes than other groups. CONCLUSIONS: According to the findings, injecting MSCs intraventricularly versus cell-conditioned medium can be a more effective approach to improving chronic demyelination in degenerative diseases like MS.


Cuprizone , Demyelinating Diseases , Disease Models, Animal , Inflammation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice, Inbred C57BL , Animals , Mesenchymal Stem Cell Transplantation/methods , Mice , Mesenchymal Stem Cells/metabolism , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Culture Media, Conditioned/pharmacology , Inflammation/pathology , Inflammation/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Oligodendroglia/metabolism , Remyelination , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Multiple Sclerosis/metabolism , Multiple Sclerosis/chemically induced , Tumor Necrosis Factor-alpha/metabolism , Male , Myelin Sheath/metabolism
6.
Int J Mol Sci ; 25(10)2024 May 10.
Article En | MEDLINE | ID: mdl-38791236

The conditioned medium (CM) obtained from mesenchymal stromal cell (MSC) culture has excellent cell growth-promoting activity and is used for cosmetics and healthcare products. Unlike pharmaceuticals, strict efficacy verification is not legally required for these products. However, their efficacy must be substantiated as commercial products. We attempted to simplify CM production and to standardize the evaluation of the growth-promoting activity of CM. CM was obtained through the culturing of two lines of commercially available human adipose tissue-derived MSCs using MEMα with or without 10% fetal bovine serum (FBS) for 24 h. Non-CM control media were produced by the same protocol without MSCs. Growth-promoting activities of the CM were estimated by [3H]-thymidine pulse. CM were subjected to molecular weight fractionation with ultrafiltration using 10 k-, 30 k-, 50 k-, and 100 k-membranes. The FBS-free CMs showed 1.34- to 1.85-fold increases and FBS-containing CMs showed 1.45- to 1.67-fold increases in proliferation-promoting activity compared with non-CM controls, regardless of the source of the cell. The thymidine incorporation levels were approximately three times higher in FBS-containing CMs. Aged cells also showed 1.67- to 2.48-fold increases in the activity due to FBS-containing CM, but not to FBS-free CM. The CM activities were sustained even after 1 year at 4 °C. Molecular weight fractionation showed that the activity was recovered in the fraction above 100 k. Clear and stable cell-growth-promoting activity was confirmed with CMs of commercially available adipose tissue MSCs. The activity was detected in the fraction over 100 k. We propose here the importance of standardizing the production and evaluation of CMs to indicate their specific action.


Adipose Tissue , Cell Proliferation , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Culture Media, Conditioned/pharmacology , Cells, Cultured , Cell Culture Techniques/methods , Cell Culture Techniques/standards
7.
Int J Mol Sci ; 25(10)2024 May 14.
Article En | MEDLINE | ID: mdl-38791371

The process of aging is intimately linked to alterations at the tissue and cellular levels. Currently, the role of senescent cells in the tissue microenvironment is still being investigated. Despite common characteristics, different cell populations undergo distinctive morphofunctional changes during senescence. Mesenchymal stem cells (MSCs) play a pivotal role in maintaining tissue homeostasis. A multitude of studies have examined alterations in the cytokine profile that determine their regulatory function. The extracellular matrix (ECM) of MSCs is a less studied aspect of their biology. It has been shown to modulate the activity of neighboring cells. Therefore, investigating age-related changes in the MSC matrisome is crucial for understanding the mechanisms of tissue niche ageing. This study conducted a broad proteomic analysis of the matrisome of separated fractions of senescent MSCs, including the ECM, conditioned medium (CM), and cell lysate. This is the first time such an analysis has been conducted. It has been established that there is a shift in production towards regulatory molecules and a significant downregulation of the main structural and adhesion proteins of the ECM, particularly collagens, fibulins, and fibrilins. Additionally, a decrease in the levels of cathepsins, galectins, S100 proteins, and other proteins with cytoprotective, anti-inflammatory, and antifibrotic properties has been observed. However, the level of inflammatory proteins and regulators of profibrotic pathways increases. Additionally, there is an upregulation of proteins that can directly cause prosenescent effects on microenvironmental cells (SERPINE1, THBS1, and GDF15). These changes confirm that senescent MSCs can have a negative impact on other cells in the tissue niche, not only through cytokine signals but also through the remodeled ECM.


Cellular Senescence , Extracellular Matrix , Mesenchymal Stem Cells , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Humans , Extracellular Matrix/metabolism , Proteomics/methods , Proteome/metabolism , Extracellular Matrix Proteins/metabolism , Cells, Cultured , Culture Media, Conditioned/pharmacology
8.
Sci Rep ; 14(1): 10243, 2024 05 03.
Article En | MEDLINE | ID: mdl-38702388

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.


Freeze Drying , Mesenchymal Stem Cells , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Humans , Secretome/metabolism , Trehalose/metabolism , Trehalose/pharmacology , Cytokines/metabolism , Cells, Cultured , Culture Media, Conditioned/chemistry , Cryopreservation/methods , Temperature
9.
Am J Reprod Immunol ; 91(5): e13853, 2024 May.
Article En | MEDLINE | ID: mdl-38706383

BACKGROUND: The amniotic membrane (AM) has shown immense potential in repairing wounds due to its great regenerative qualities. Although the role of AM as a biological scaffold in repairing wounds has been studied well, the tissue regenerative potential of AM-derived mesenchymal stem cells (MSCs) and conditioned media (CM) derived from it remains to be discovered as of now. Here, we examined the wound healing abilities of fresh and frozen thawed rabbit AM (rAM) along with the MSCs and their lyophilised CM in rabbits challenged with skin wounds. METHODS: To elucidate the role of rAM-MSCs and its CM in repairing the wound, we isolated it from the freshly derived placenta and characterised their differentiation potential by performing an in vitro tri-lineage differentiation assay besides other standard confirmations. We compared the wound repair capacities of rAM-MSCs and lyophilised CM with the fresh and cryopreserved AM at different timelines by applying them to excision wounds created in rabbits. RESULTS: By monitoring wound contractions and tissue histology of wounded skin at different time points after the application, we observed that rAM-MSCs and rAM-MSC-derived CM significantly promoted wound closure compared to the control group. We also observed that the wound closure capacity of rAM-MSCs and rAM-MSC-derived CM is as efficient as fresh and cryopreserved rAM. CONCLUSION: Our findings suggest that rAM-MSCs and rAM-MSC derived CM can be effectively used to treat skin wounds in animals and correctly delivered to the damaged tissue using AM as a bioscaffold, either fresh or frozen.


Amnion , Mesenchymal Stem Cells , Wound Healing , Animals , Rabbits , Female , Mesenchymal Stem Cells/cytology , Cell Differentiation , Culture Media, Conditioned/pharmacology , Mesenchymal Stem Cell Transplantation/methods , Skin/injuries , Skin/pathology , Pregnancy , Disease Models, Animal , Cells, Cultured , Transplantation, Homologous
10.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38732109

Adipose-derived mesenchymal stem cells (ASCs) are adult multipotent stem cells, able to differentiate toward neural elements other than cells of mesodermal lineage. The aim of this research was to test ASC neural differentiation using melatonin combined with conditioned media (CM) from glial cells. Isolated from the lipoaspirate of healthy donors, ASCs were expanded in a basal growth medium before undergoing neural differentiation procedures. For this purpose, CM obtained from olfactory ensheathing cells and from Schwann cells were used. In some samples, 1 µM of melatonin was added. After 1 and 7 days of culture, cells were studied using immunocytochemistry and flow cytometry to evaluate neural marker expression (Nestin, MAP2, Synapsin I, GFAP) under different conditions. The results confirmed that a successful neural differentiation was achieved by glial CM, whereas the addition of melatonin alone did not induce appreciable changes. When melatonin was combined with CM, ASC neural differentiation was enhanced, as demonstrated by a further improvement of neuronal marker expression, whereas glial differentiation was attenuated. A dynamic modulation was also observed, testing the expression of melatonin receptors. In conclusion, our data suggest that melatonin's neurogenic differentiation ability can be usefully exploited to obtain neuronal-like differentiated ASCs for potential therapeutic strategies.


Cell Differentiation , Melatonin , Mesenchymal Stem Cells , Melatonin/pharmacology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Humans , Cell Differentiation/drug effects , Cells, Cultured , Adipose Tissue/cytology , Neurons/cytology , Neurons/metabolism , Neurons/drug effects , Culture Media, Conditioned/pharmacology , Schwann Cells/cytology , Schwann Cells/metabolism , Schwann Cells/drug effects , Neurogenesis/drug effects , Adult , Nestin/metabolism , Nestin/genetics , Glial Fibrillary Acidic Protein/metabolism , Neuroglia/drug effects , Neuroglia/cytology , Neuroglia/metabolism , Synapsins/metabolism
11.
Nutrients ; 16(9)2024 Apr 26.
Article En | MEDLINE | ID: mdl-38732549

Oleocanthal (OC) is a monophenol of extra-virgin olive oil (EVOO) endowed with antibiotic, cardioprotective and anticancer effects, among others, mainly in view of its antioxidant and anti-inflammatory properties. OC has been largely investigated in terms of its anticancer activity, in Alzheimer disease and in collagen-induced arthritis; however, the possibility that it can also affect muscle biology has been totally overlooked so far. This study is the first to describe that OC modulates alterations induced in C2C12 myotubes by stimuli known to induce muscle wasting in vivo, namely TNF-α, or in the medium conditioned by the C26 cachexia-inducing tumor (CM-C26). C2C12 myotubes were exposed to CM-C26 or TNF-α in the presence or absence of OC for 24 and 48 h and analyzed by immunofluorescence and Western blotting. In combination with TNF-α or CM-C26, OC was revealed to be able to restore both the myotube's original size and morphology and normal levels of both atrogin-1 and MuRF1. OC seems unable to impinge on the autophagic-lysosomal proteolytic system or protein synthesis. Modulations towards normal levels of the expression of molecules involved in myogenesis, such as Pax7, myogenin and MyHC, were also observed in the myotube cultures exposed to OC and TNF-α or CM-C26. In conclusion, the data presented here show that OC exerts a protective action in C2C12 myotubes exposed to TNF-α or CM-C26, with mechanisms likely involving the downregulation of ubiquitin-proteasome-dependent proteolysis and the partial relief of myogenic differentiation impairment.


Catechols , Cyclopentane Monoterpenes , Muscle Fibers, Skeletal , Muscle Proteins , Muscular Atrophy , Tumor Necrosis Factor-alpha , Animals , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Mice , Tumor Necrosis Factor-alpha/metabolism , Muscular Atrophy/prevention & control , Muscular Atrophy/metabolism , Muscle Proteins/metabolism , Cyclopentane Monoterpenes/pharmacology , Catechols/pharmacology , Cell Line , SKP Cullin F-Box Protein Ligases/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Muscle Development/drug effects , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Autophagy/drug effects , Phenols/pharmacology , Cachexia/prevention & control , Culture Media, Conditioned/pharmacology , Aldehydes
12.
Endocrinology ; 165(6)2024 Apr 29.
Article En | MEDLINE | ID: mdl-38712392

Long-term ß-adrenoceptor (ß-AR) stimulation is a pathological mechanism associated with cardiovascular diseases resulting in endothelial and perivascular adipose tissue (PVAT) dysfunction. In this study, we aimed to identify whether ß-adrenergic signaling has a direct effect on PVAT. Thoracic aorta PVAT was obtained from male Wistar rats and cultured ex vivo with the ß-AR agonist isoproterenol (Iso; 1 µM) or vehicle for 24 hours. Conditioned culture medium (CCM) from Iso-treated PVAT induced a marked increase in aorta contractile response, induced oxidative stress, and reduced nitric oxide production in PVAT compared to vehicle. In addition, Iso-treated PVAT and PVAT-derived differentiated adipocytes exhibited higher corticosterone release and protein expression of 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1), an enzyme responsible for de novo synthesis of corticosterone. Macrophages exposed to Iso also exhibited increased corticosterone release in response to ß-AR stimulation. Incubation of Iso-treated PVAT and PVAT-derived differentiated adipocytes with ß3-AR antagonist restored aorta contractile function modulated by Iso-CCM and normalized 11ß-HSD1 protein expression. These results show that ß3-AR signaling leads to upregulation of 11ß-HSD1 in PVAT, thus increasing corticosterone release and contributing to impair the anticontractile function of this tissue.


11-beta-Hydroxysteroid Dehydrogenase Type 1 , Corticosterone , Isoproterenol , Rats, Wistar , Animals , Male , Rats , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Isoproterenol/pharmacology , Corticosterone/metabolism , Adrenergic beta-Agonists/pharmacology , Adipose Tissue/metabolism , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Adipocytes/metabolism , Adipocytes/drug effects , Receptors, Adrenergic, beta/metabolism , Oxidative Stress/drug effects , Nitric Oxide/metabolism , Culture Media, Conditioned/pharmacology
13.
Eur J Med Res ; 29(1): 270, 2024 May 04.
Article En | MEDLINE | ID: mdl-38704575

BACKGROUND: This study aims to investigate the effects of a conditioned medium (CM) from human umbilical cord mesenchymal stem cells (HuMSCs) cultivated in gelatin sponge (GS-HuMSCs-CM) on hair growth in a mouse model. METHODS: CM was collected from the HuMSCs cultivated in a monolayer or in a gelatin sponge. Vascular endothelial growth factor (VEGF), insulin-like growth factor-1 (IGF-1), keratinocyte growth factor (KGF), and hepatocyte growth factor (HGF) levels in CMs were measured by enzyme-linked immunosorbent assays (ELISAs). A hair loss model by a C57 BL/6J mouse was prepared. The effects of GS-HuMSCs-CM and HuMSCs on hair regrowth in mice were investigated by intradermal injection in the depilated back skin with normal saline (NS) as the control. The time for hair regrowth and full covering in depilated areas was observed, and the hair growth was evaluated histologically and by grossly measuring hair length and diameter. RESULTS: Compared with monolayer cultured cells, the three-dimensional (3D) culture of HuMSCs in gelatin sponge drastically increased VEGF, IGF-1, KGF, and HGF production. GS-HuMSCs-CM and HuMSCs injection both promoted hair regeneration in mice, while GS-HuMSCs-CM presented more enhanced effects in hair length, hair diameter, and growth rate. GS-HuMSCs-CM significantly promoted angiogenesis in injected skin areas, which might also contribute to faster hair regrowth. CONCLUSION: GS-HuMSCs-CM exerted significant effects on inducing hair growth and promoted skin angiogenesis in C57BL/6J mice.


Hair , Insulin-Like Growth Factor I , Mesenchymal Stem Cells , Umbilical Cord , Animals , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Humans , Culture Media, Conditioned/pharmacology , Mice , Umbilical Cord/cytology , Hair/growth & development , Hair/drug effects , Insulin-Like Growth Factor I/metabolism , Vascular Endothelial Growth Factor A/metabolism , Hepatocyte Growth Factor/metabolism , Gelatin/chemistry , Tissue Scaffolds/chemistry , Mice, Inbred C57BL , Cells, Cultured , Fibroblast Growth Factor 7/metabolism
14.
Ann Plast Surg ; 92(4S Suppl 2): S207-S209, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38556675

INTRODUCTION: Autologous fat grafting (AFG) is a common technique used to enhance aesthetic outcomes in postmastectomy breast reconstruction patients. Adipokines are hormones secreted by adipose tissue that play a critical role in regulating metabolic processes and the immune system. However, dysregulated adipokine secretion and signaling can contribute to the development and progression of cancer by promoting angiogenesis, altering the immune response, and inducing the epithelial mesenchymal transition. We aimed to assess how breast cancer cells behave in conditioned media derived from fat grafting lipoaspirates and gain a better understanding of the potential interactions that may occur within the tumor microenvironment. METHODS: Patients who were undergoing AFG as a part of breast reconstruction at NY-Presbyterian/Weill Cornell Medical Center between March 2021 and July 2023 were consented and enrolled in the study. This study was approved by the Weill Cornell Medicine Institutional Review Board (#20-10022850-14). Conditioned media is created using 20% of patient lipoaspirate secretome and 80% starving media. The growth of MCF-7, a human ER/PR+ breast cancer cell line, in conditioned media is assessed using CyQUANT. RESULTS: The breast cancer cells incubated in conditioned media displayed similar growth trends as those in complete media, which is enriched for cell growth (P > 0.05). MCF-7 cell behavior in conditioned media differed significantly from their proliferation patterns when serum starved in 100% starving media (P < 0.05). DISCUSSION: Our results suggest that there may be inherent factors within the lipoaspirate that may promote MCF-7 proliferation. One potential implication is that AFG used for breast reconstruction should be delayed until local-regional disease control has been established. In addition, based on the in vitro proliferation patterns of breast cancer cells in conditioned media, the safety profile of AFG may be enhanced if the procedure is performed after attaining negative margins and the completion breast cancer treatment.


Breast Neoplasms , Humans , Female , Breast Neoplasms/surgery , MCF-7 Cells , Culture Media, Conditioned/pharmacology , Mastectomy , Cell Proliferation , Adipose Tissue/transplantation , Tumor Microenvironment
15.
Cell Mol Life Sci ; 81(1): 180, 2024 Apr 13.
Article En | MEDLINE | ID: mdl-38613672

Aberrant remodeling of uterine spiral arteries (SPA) is strongly associated with the pathogenesis of early-onset preeclampsia (EOPE). However, the complexities of SPA transformation remain inadequately understood. We conducted a single-cell RNA sequencing analysis of whole placental tissues derived from patients with EOPE and their corresponding controls, identified DAB2 as a key gene of interest and explored the mechanism underlying the communication between Extravillous trophoblast cells (EVTs) and decidual vascular smooth muscle cells (dVSMC) through cell models and a placenta-decidua coculture (PDC) model in vitro. DAB2 enhanced the motility and viability of HTR-8/SVneo cells. After exposure to conditioned medium (CM) from HTR-8/SVneoshNC cells, hVSMCs exhibited a rounded morphology, indicative of dedifferentiation, while CM-HTR-8/SVneoshDAB2 cells displayed a spindle-like morphology. Furthermore, the PDC model demonstrated that CM-HTR-8/SVneoshDAB2 was less conducive to vascular remodeling. Further in-depth mechanistic investigations revealed that C-X-C motif chemokine ligand 8 (CXCL8, also known as IL8) is a pivotal regulator governing the dedifferentiation of dVSMC. DAB2 expression in EVTs is critical for orchestrating the phenotypic transition and motility of dVSMC. These processes may be intricately linked to the CXCL8/PI3K/AKT pathway, underscoring its central role in intricate SPA remodeling.


Eosine Yellowish-(YS)/analogs & derivatives , Interleukin-8 , Phosphatidylethanolamines , Pre-Eclampsia , Pregnancy , Humans , Female , Interleukin-8/genetics , Phosphatidylinositol 3-Kinases , Pre-Eclampsia/genetics , Placenta , Arteries , Culture Media, Conditioned , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins
16.
Arq Bras Oftalmol ; 87(2): e2022, 2024.
Article En | MEDLINE | ID: mdl-38655938

PURPOSES: To determine the best protocol in obtaining the higher yield of conditioned culture medium to be used for the bone marrow mesenchymal stem cell differentiation into corneal epithelial cells, five techniques for the primary culture of human corneal epithelial cells were evaluated. METHODS: The studied culture techniques of corneal epithelial cells were: explants in culture flasks with and without hydrophilic surface treatment, on amniotic membrane, with enzymatic digestion, and by corneal scraping. The conditioned culture medium collected from these cultures was used to differentiate human bone marrow mesenchymal stem cells into corneal epithelial cells, which were characterized using flow cytometry with pan-cytokeratin and the corneal-specific markers, cytokeratin 3 and cytokeratin 12. RESULTS: The culture technique using flasks with hydrophilic surface treatment resulted in the highest yield of conditioned culture medium. Flasks without surface treatment resulted to a very low success rate. Enzymatic digestion and corneal scraping showed contamination with corneal fibroblasts. The culture on amniotic membranes only allowed the collection of culture medium during the 1st cell confluence. The effectiveness of cell differentiation was confirmed by cytometry analysis using the collected conditioned culture medium, as demonstrated by the expressions of cytokeratin 3 (95.3%), cytokeratin 12 (93.4%), and pan-cytokeratin (95.3%). CONCLUSION: The culture of corneal epithelial cell explants in flasks with hydrophilic surface treatment is the best technique for collecting a higher yield of conditioned culture medium to be used to differentiate mesenchymal stem cells.


Cell Culture Techniques , Cell Differentiation , Epithelium, Corneal , Flow Cytometry , Mesenchymal Stem Cells , Humans , Culture Media, Conditioned , Epithelium, Corneal/cytology , Cell Differentiation/physiology , Flow Cytometry/methods , Mesenchymal Stem Cells/cytology , Cell Culture Techniques/methods , Amnion/cytology , Cells, Cultured , Keratin-3/metabolism , Keratin-3/analysis , Keratin-12/metabolism , Reproducibility of Results
17.
J Neurosci Res ; 102(4): e25334, 2024 Apr.
Article En | MEDLINE | ID: mdl-38656648

Iron deficiency (ID) has been shown to affect central nervous system (CNS) development and induce hypomyelination. Previous work from our laboratory in a gestational ID model showed that both oligodendrocyte (OLG) and astrocyte (AST) maturation was impaired. To explore the contribution of AST iron to the myelination process, we generated an in vitro ID model by silencing divalent metal transporter 1 (DMT1) in AST (siDMT1 AST) or treating AST with Fe3+ chelator deferoxamine (DFX; DFX AST). siDMT1 AST showed no changes in proliferation but remained immature. Co-cultures of oligodendrocyte precursors cells (OPC) with siDMT1 AST and OPC cultures incubated with siDMT1 AST-conditioned media (ACM) rendered a reduction in OPC maturation. These findings correlated with a decrease in the expression of AST-secreted factors IGF-1, NRG-1, and LIF, known to promote OPC differentiation. siDMT1 AST also displayed increased mitochondrial number and reduced mitochondrial size as compared to control cells. DFX AST also remained immature and DFX AST-conditioned media also hampered OPC maturation in culture, in keeping with a decrease in the expression of AST-secreted growth factors IGF-1, NRG-1, LIF, and CNTF. DFX AST mitochondrial morphology and number showed results similar to those observed in siDMT1 AST. In sum, our results show that ID, induced through two different methods, impacts AST maturation and mitochondrial functioning, which in turn hampers OPC differentiation.


Astrocytes , Cell Differentiation , Iron Deficiencies , Oligodendroglia , Astrocytes/metabolism , Astrocytes/drug effects , Oligodendroglia/metabolism , Oligodendroglia/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cation Transport Proteins/metabolism , Coculture Techniques , Culture Media, Conditioned/pharmacology , Rats , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Deferoxamine/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Iron/metabolism
18.
Mol Biol Rep ; 51(1): 570, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658405

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.


Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Spinal Cord Injuries , Transcriptome , Umbilical Cord , Spinal Cord Injuries/therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/genetics , Mesenchymal Stem Cells/metabolism , Culture Media, Conditioned/pharmacology , Umbilical Cord/cytology , Umbilical Cord/metabolism , Humans , Animals , Mesenchymal Stem Cell Transplantation/methods , Transcriptome/genetics , Rats , Secretome/metabolism , Cell Differentiation , Neurons/metabolism , Disease Models, Animal , Interleukin-10/genetics , Interleukin-10/metabolism , Cells, Cultured , Proteomics/methods
19.
Biomed Mater ; 19(3)2024 Apr 25.
Article En | MEDLINE | ID: mdl-38604155

The standard surgical procedure for abdominal hernia repair with conventional prosthetic mesh still results in a high recurrence rate. In the present study, we propose a fibroblast matrix implant (FMI), which is a three-dimensional (3D) poly-L-lactic acid scaffold coated with collagen (matrix) and seeded with fibroblasts, as an alternative mesh for hernia repair. The matrix was seeded with fibroblasts (cellularized) and treated with a conditioned medium (CM) of human Umbilical Cord Mesenchymal Stem Cells (hUC-MSC). Fibroblast proliferation and function were assessed and compared between treated with CM hUC-MSC and untreated group, 24 h after seeding onto the matrix (n= 3). To study the matricesin vivo,the hernia was surgically created on male Sprague Dawley rats and repaired with four different grafts (n= 3), including a commercial mesh (mesh group), a matrix without cells (cell-free group), a matrix seeded with fibroblasts (FMI group), and a matrix seeded with fibroblasts and cultured in medium treated with 1% CM hUC-MSC (FMI-CM group).In vitroexamination showed that the fibroblasts' proliferation on the matrices (treated group) did not differ significantly compared to the untreated group. CM hUC-MSC was able to promote the collagen synthesis of the fibroblasts, resulting in a higher collagen concentration compared to the untreated group. Furthermore, thein vivostudy showed that the matrices allowed fibroblast growth and supported cell functionality for at least 1 month after implantation. The highest number of fibroblasts was observed in the FMI group at the 14 d endpoint, but at the 28 d endpoint, the FMI-CM group had the highest. Collagen deposition area and neovascularization at the implantation site were observed in all groups without any significant difference between the groups. FMI combined with CM hUC-MSC may serve as a better option for hernia repair, providing additional reinforcement which in turn should reduce hernia recurrence.


Cell Proliferation , Collagen , Fibroblasts , Herniorrhaphy , Incisional Hernia , Mesenchymal Stem Cells , Rats, Sprague-Dawley , Surgical Mesh , Tissue Scaffolds , Animals , Fibroblasts/metabolism , Rats , Male , Humans , Mesenchymal Stem Cells/cytology , Herniorrhaphy/methods , Herniorrhaphy/instrumentation , Collagen/chemistry , Tissue Scaffolds/chemistry , Incisional Hernia/surgery , Polyesters/chemistry , Materials Testing , Culture Media, Conditioned/pharmacology , Biocompatible Materials/chemistry , Cells, Cultured , Hernia, Abdominal/surgery , Umbilical Cord/cytology
20.
Biol Pharm Bull ; 47(4): 809-817, 2024.
Article En | MEDLINE | ID: mdl-38583954

Several reports assume that myocardial necroptotic cell death is induced during the development of chronic heart failure. Although it is well accepted that angiotensin II induces apoptotic cell death of cardiac myocytes, the involvement of angiotensin II in the induction of myocardial necroptosis during the development of heart failure is still unknown. Therefore, we examined the role of angiotensin II in myocardial necroptosis using rat failing hearts following myocardial infarction and cultured cardiomyocytes. We found that administration of azilsartan, an angiotensin II AT1 receptor blocker, or trandolapril, an angiotensin-converting enzyme inhibitor, to rats from the 2nd to the 8th week after myocardial infarction resulted in preservation of cardiac function and attenuation of mixed lineage kinase domain-like (MLKL) activation. Furthermore, the ratio of necroptotic cell death was increased in neonatal rat ventricular cardiomyocytes cultured with conditioned medium from rat cardiac fibroblasts in the presence of angiotensin II. This increase in necroptotic cells was attenuated by pretreatment with azilsartan. Furthermore, activated MLKL was increased in cardiomyocytes cultured in conditioned medium. Pretreatment with azilsartan also prevented the conditioned medium-induced increase in activated MLKL. These results suggest that angiotensin II contributes to the induction of myocardial necroptosis during the development of heart failure.


Heart Failure , Myocardial Infarction , Animals , Rats , Angiotensin II/pharmacology , Angiotensin II/metabolism , Angiotensin Receptor Antagonists , Culture Media, Conditioned/metabolism , Heart Failure/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac , Protein Kinases/metabolism
...