Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Microsc Res Tech ; 87(7): 1598-1614, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38441397

ABSTRACT

Cardiotoxicity induced by doxorubicin (Dox) is a major complication in cancer patients. Exosomes (Ex) derived from mesenchymal cells could be a promising therapeutic for various heart diseases. This study investigated the role of Ex in Dox-induced cardiotoxicity and its mechanistic insights, using Sacubitril/valsartan (S/V) as a reference drug widely recommended in heart failure management. The study involved 24 Wistar rats, divided into a control, Dox, Dox + S/V, and Dox + Ex groups. The rats were assessed for cardiac enzymes, inflammatory and oxidative stress markers. Immunohistochemical expression of caspase-1, nuclear factor erythroid 2-related factor 2 (NrF2), E-Cadherin, CD117/c-kit, and Platelet-derived growth factor-α (PDGFα) was evaluated. P53 and Annexin V were assessed by PCR. Histological examination was performed using hematoxylin and eosin and Sirius red stains. Ex ameliorated the adverse cardiac pathological changes and significantly decreased the cardiac enzymes and inflammatory and oxidative stress markers. Ex also exerted antifibrotic and antiapoptotic effect in heart tissue. Ex treatment also improved NrF2 immunohistochemistry, up-regulated E-Cadherin immune expression, and restored the telocyte markers CD117/c-kit and PDGFα. Ex can mitigate Dox-induced cardiotoxicity by acting as an anti-inflammatory, antioxidant, antiapoptotic, and antifibrotic agents, restoring telocytes and modulating epithelial mesenchymal transition. RESEARCH HIGHLIGHTS: Exosomes exhibit positive expression for CD90 and CD105 whereas showing -ve expression for CD 34 by flow cytometry. Exosomes restore the immunohistochemical expression of the telocytes markers CD117/c-kit and PDGFα. Exosomes alleviate myocardial apoptosis, oxidative stress and fibrosis.


Subject(s)
Apoptosis , Cardiotoxicity , Doxorubicin , Exosomes , Fibrosis , Inflammation , Mesenchymal Stem Cells , Oxidative Stress , Rats, Wistar , Telocytes , Animals , Doxorubicin/adverse effects , Doxorubicin/toxicity , Exosomes/metabolism , Exosomes/drug effects , Apoptosis/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Rats , Oxidative Stress/drug effects , Telocytes/drug effects , Male , Myocardium/pathology
2.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Article in English | MEDLINE | ID: mdl-33558238

ABSTRACT

Propranolol, a nonselective ß-adrenergic receptor (ADRB) antagonist, is the first-line therapy for severe infantile hemangiomas (IH). Since the incidental discovery of propranolol efficacy in IH, preclinical and clinical investigations have shown evidence of adjuvant propranolol response in some malignant tumors. However, the mechanism for propranolol antitumor effect is still largely unknown, owing to the absence of a tumor model responsive to propranolol at nontoxic concentrations. Immunodeficient mice engrafted with different human tumor cell lines were treated with anti-VEGF bevacizumab to create a model sensitive to propranolol. Proteomics analysis was used to reveal propranolol-mediated protein alteration correlating with tumor growth inhibition, and Aquaporin-1 (AQP1), a water channel modulated in tumor cell migration and invasion, was identified. IH tissues and cells were then functionally investigated. Our functional protein association networks analysis and knockdown of ADRB2 and AQP1 indicated that propranolol treatment and AQP1 down-regulation trigger the same pathway, suggesting that AQP1 is a major driver of beta-blocker antitumor response. Examining AQP1 in human hemangioma samples, we found it exclusively in a perivascular layer, so far unrecognized in IH, made of telocytes (TCs). Functional in vitro studies showed that AQP1-positive TCs play a critical role in IH response to propranolol and that modulation of AQP1 in IH-TC by propranolol or shAQP1 decreases capillary-like tube formation in a Matrigel-based angiogenesis assay. We conclude that IH sensitivity to propranolol may rely, at least in part, on a cross talk between lesional vascular cells and stromal TCs.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Aquaporin 1/metabolism , Hemangioma, Capillary/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Neovascularization, Pathologic/metabolism , Propranolol/pharmacology , Telocytes/metabolism , Animals , Cell Line, Tumor , Cell Movement , Hemangioma, Capillary/drug therapy , Humans , Mice , Neoplastic Syndromes, Hereditary/drug therapy , Neovascularization, Pathologic/drug therapy , Propranolol/therapeutic use , Proteome/genetics , Proteome/metabolism , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Telocytes/drug effects , Telocytes/physiology
3.
Sci Rep ; 11(1): 872, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441634

ABSTRACT

Dendritic cells (DCs) are innate immune cells which engulf, process and present antigens to the naïve T-lymphocyte cells. However, little is known about the effect of melatonin on the DCs. The present study aimed to investigate the morphology and distribution of the DCs by transmission electron microscopy and Immunohistochemistry after melatonin administration. A total of 8 out of 15 adult ram was randomly selected to receive the melatonin implant and the remaining 7 animals received melatonin free implants. DCs showed positive immunoreactivity for CD117, S-100 protein and CD34. There is an obvious increase in the number of the positive immunoreactive cells to CD3, estrogen receptor alpha and progesterone in the treated groups. The expression of CD56 and MHCII in the DCs was abundant in the treated groups. The ultrastructure study revealed that melatonin exerts a stimulatory effect on the DCs which was associated with increment in the secretory activity of DCs. The secretory activity demarcated by an obvious increase in the number of mitochondria, cisternae of rER and a well-developed Golgi apparatus. The endosomal- lysosomal system was more developed in the treated groups. A rod-shaped Birbeck granule was demonstrated in the cytoplasm of the melatonin treated group. DCs were observed in a close contact to telocytes, T-Lymphocytes, nerve fibers and blood vessels. Taken together, melatonin administration elicits a stimulatory action on the DCs and macrophages through increasing the size, the number and the endosomal compartments which may correlate to increased immunity.


Subject(s)
Dendritic Cells, Follicular/metabolism , Melatonin/pharmacology , Seminal Vesicles/metabolism , Animals , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dendritic Cells, Follicular/drug effects , Lymphocytes/immunology , Macrophages/drug effects , Macrophages/metabolism , Male , Melatonin/metabolism , Seminal Vesicles/drug effects , Sheep , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Telocytes/drug effects , Telocytes/immunology
4.
Folia Med Cracov ; 60(2): 81-95, 2020 09 28.
Article in English | MEDLINE | ID: mdl-33252597

ABSTRACT

BACKGROUND: Uterine leiomyoma is the most widespread benign tumor affecting women of childbearing age. There are still gaps in the understanding of its pathogenesis. Telocytes are unique cells found in more than 50 different locations inside the human body. The functional relationship between cells could clarify the pathogenesis of leiomyomata. Examination of membrane receptors on telocytes could explain their role in fibrosis, oxidative stress, and myometrial contractility. AIM: This research was conducted to assess the density of telocytes in terms of their putative role in leiomyoma formation by focusing on their correlation with the expression of estrogen and progesterone receptors. METHODS: For gross evaluation of uterine tissue samples from leiomyoma, routine histology of adjacent and unaffected myometrium was performed. Immunohistochemical analysis of c-kit, tryptase, CD34, PDGFRα (telocyte-specific), and ER and PRs (estrogen and progesterone receptors) was performed to examine uterine telocytes and the expression of sex steroid receptors. RESULTS: The decline in telocyte density in leiomyoma foci was correlated with high progesterone expression and low estrogen receptor expression. The unchanged myometrium showed the opposite correlation and balance between both steroid hormone receptors. The difference in sex steroid receptor expression is correlated with the density of uterine telocytes, which emphasizes their conductor function. CONCLUSIONS: A reduction in telocyte density and the changes in examined marker expression demonstrate the involvement of telocytes in local homeostasis. The expression of membrane receptors explicitly indicates their functional potential in the human myometrium, focusing attention on contractility and local homeostasis.


Subject(s)
Estradiol Congeners/pharmacokinetics , Leiomyoma/diagnostic imaging , Leiomyoma/pathology , Progesterone/pharmacokinetics , Telocytes/drug effects , Telocytes/physiology , Uterine Neoplasms/diagnostic imaging , Uterine Neoplasms/pathology , Adult , Aged , Estradiol Congeners/physiology , Female , Humans , Middle Aged , Poland , Progesterone/physiology
5.
Neurotoxicology ; 77: 193-204, 2020 03.
Article in English | MEDLINE | ID: mdl-32007490

ABSTRACT

Given the well-known antioxidant and neuroprotective properties of quercetin, the aim of this work was to evaluate the effects of quercetin stabilized by microencapsulation at two doses (10 mg kg-1 and 100 mg kg-1) on the oxidative/antioxidant status, number and morphological features of ICC, nitrergic neurons and M2-like macrophages in jejunum of diabetic rats. The rats were randomly distributed into six groups: normoglycemic control (N), diabetic control (D) and either normoglycemic or diabetic groups treated with quercetin-loaded microcapsules at a dose of 10 mg kg-1 (NQ10 and DQ10, respectively) or 100 mg kg-1 (NQ100 and DQ100, respectively). After 60 days, the jejunum was collected. Whole mounts were immunostained for Ano1, nNOS and CD206, and oxidative stress levels and total antioxidant capacity of the jejunum were measured. Diabetes led to a loss of ICC and nitrergic neurons, but increased numbers of M2-like macrophages and elevated levels of oxidative stress were seen in diabetic animals. High-dose administration of quercetin (100 mg kg-1) further aggravated the diabetic condition (DQ100) but this treatment resulted in harmful effects on healthy rats (NQ100), pointing to a pro-oxidant activity. However, low-dose administration of quercetin (10 mg kg-1) gave rise to antioxidant and protective effects on ICC, nNOS, macrophages and oxidative/antioxidant status in DQ100, but NQ100 displayed infrequent negative outcomes in normoglycemic animals. Microencapsulation of the quercetin may become promising alternatives to reduce diabetes-induced oxidative stress but antioxidant therapies should be careful used under healthy status to avoid toxic effects.


Subject(s)
Antioxidants/administration & dosage , Diabetes Mellitus, Type 1/metabolism , Jejunum/drug effects , Macrophages/drug effects , Nitrergic Neurons/drug effects , Quercetin/administration & dosage , Telocytes/drug effects , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/chemically induced , Drug Compounding , Jejunum/metabolism , Macrophages/metabolism , Male , Myenteric Plexus/drug effects , Myenteric Plexus/metabolism , Nitrergic Neurons/metabolism , Oxidative Stress/drug effects , Rats, Wistar , Streptozocin/administration & dosage , Telocytes/metabolism
6.
Protoplasma ; 257(2): 353-369, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31637525

ABSTRACT

The adrenal glands of 15 adult Soay rams were used to study the effect of melatonin on their vascular elements and cellular organization. A significant increase in the cross-sectional area of the blood sinusoids was demonstrated after melatonin administration. The vimentin-expressing mesenchymal cells were increased in the melatonin-treated group. Intensive S-100 protein expression was observed in the sustentacular cells and telocytes (TCs) of the treated groups. Moreover, S-100 protein expressed intensively in the dendritic cells that distributed around the blood sinusoids. Dendritic cells showed positive immunoreactivity for CD8 and CD103. Many dendritic cells with well-defined processes were observed close to the nerve fibers after melatonin administration. A significant increase in the number and diameter of dendritic cells after melatonin treatment was demonstrated. Many highly active TCs were observed in the medulla of the treated group, which were characterized by long telopodes (Tps) containing abundant secretory vesicles that released into the extracellular milieu and towards the dendritic cells. In the melatonin-treated groups, the nerve fibers showed a significant increase in their cross-sectional area accompanied by an increase in the activity of Schwann cells and neighboring dendritic cells. In the treated group, TCs and DCs appear to contribute to angiogenesis. A planner contact between Tps and the stem cell was demonstrated in the treated group. Melatonin induced a stimulatory action on the vascular and neuronal elements of the adrenal gland. Moreover, it enhances the activity of a variety of cells including telocytes, dendritic, sustentacular, and Schwann cells.


Subject(s)
Adrenal Glands/drug effects , Central Nervous System Depressants/therapeutic use , Melatonin/therapeutic use , Telocytes/drug effects , Animals , Central Nervous System Depressants/pharmacology , Male , Melatonin/pharmacology , Seasons , Sheep
7.
Sci Rep ; 9(1): 3761, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30842587

ABSTRACT

Telocytes are CD34-positive cells with a fusiform cell body and long, thin cytoplasmic projections called telopodes. These cells were detected in the stroma of various organs, including the prostate. The prostate is a complex gland capable of undergoing involution due to low testosterone levels; and this condition can be reversed with testosterone replacement. Telocyte function in the mature prostate remains to be dermined, and it is not known whether telocytes can take place in tissue remodeling during prostate involution and regrowth. The present study employed structural, ultrastructural and immunohistochemical methods to investigate the telocyte's phenotypes in the ventral prostate (VP) from control (CT), castrated (CS) and testosterone replacement (TR) groups of adult male Wistar rats. Telocytes were found in the subepithelial, perimuscular and interstitical regions around glandular acini. Telocytes from CT animals have condensed chromatin and long and thin telopodes. In CS group, telocytes appeared quiescent and exhibited layers of folded up telopodes. After TR, telocytes presented loose chromatin, abundant rough endoplasmic reticulum and enlarged telopodes, closely associated with bundles of collagen fibrils. We called these cells "telocytes with a synthetic phenotype". As testosterone levels and glandular morphology returned toward to the CT group parameters, after 10 days of TR, these telocytes progressively switched to the normal phenotype. Our results demonstrate that telocytes exhibit phenotypic plasticity upon androgen manipulation and interact with fibroblast and smooth muscle cells to maintain glandular architecture in control animals and during tissue remodeling after hormonal manipulation.


Subject(s)
Prostate/cytology , Telocytes/cytology , Testosterone Propionate/administration & dosage , Animals , Antigens, CD34/metabolism , Male , Orchiectomy , Prostate/drug effects , Prostate/growth & development , Prostate/metabolism , Rats , Rats, Wistar , Telocytes/drug effects , Telocytes/metabolism , Testosterone/blood , Testosterone Propionate/pharmacology
8.
J Transl Med ; 17(1): 431, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31888636

ABSTRACT

BACKGROUND: Telocytes (TCs) have the capacity of cell-cell communication with adjacent cells within the tissue, contributing to tissue repair and recovery from injury. The present study aims at investigating the molecular mechanisms by which the TGFß1-ITGB1-PI3K signal pathways regulate TC cycle and proliferation. METHODS: Gene expression of integrin (ITG) family were measured in mouse primary TCs to compare with other cells. TC proliferation, movement, cell cycle, and PI3K isoform protein genes were assayed in ITGB1-negative or positive mouse lung TCs treated with the inhibition of PI3Kp110α, PI3Kα/δ, PKCß, or GSK3, followed by TGFß1 treatment. RESULTS: We found the characters and interactions of ITG or PKC family member networks in primary mouse lung TCs, different from other cells in the lung tissue. The deletion of ITGB1 changed TCs sensitivity to treatment with multifunctional cytokines or signal pathway inhibitors. The compensatory mechanisms occur among TGFß1-induced PI3Kp110α, PI3Kα/δ, PKCß, or GSK3 when ITGB1 gene was deleted, leading to alterations of TC cell cycle and proliferation. Of those PI3K isoform protein genes, mRNA expression of PIK3CG altered with ITGB1-negative TC cycle and proliferation. CONCLUSION: TCs have strong capacity of proliferation through the compensatory signaling mechanisms and contribute to the development of drug resistance due to alterations of TC sensitivity.


Subject(s)
Integrin beta1/metabolism , Lung/cytology , Phosphatidylinositol 3-Kinases/metabolism , Telocytes/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antigens, CD34/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Female , Isoenzymes/metabolism , Mice, Inbred BALB C , Phosphoinositide-3 Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Telocytes/drug effects , Vimentin/metabolism
9.
Int J Mol Sci ; 19(5)2018 May 09.
Article in English | MEDLINE | ID: mdl-29747396

ABSTRACT

Voltage-gated calcium channels and estrogen receptors are essential players in uterine physiology, and their association with different calcium signaling pathways contributes to healthy and pathological conditions of the uterine myometrium. Among the properties of the various cell subtypes present in human uterine myometrium, there is increasing evidence that calcium oscillations in telocytes (TCs) contribute to contractile activity and pregnancy. Our study aimed to evaluate the effects of beta-estradiol on voltage-gated calcium channels and estrogen receptors in TCs from human uterine myometrium and to understand their role in pregnancy. For this purpose, we employed patch-clamp recordings, ratiometric Fura-2-based calcium imaging analysis, and qRT-PCR techniques for the analysis of cultured human myometrial TCs derived from pregnant and non-pregnant uterine samples. In human myometrial TCs from both non-pregnant and pregnant uterus, we evidenced by qRT-PCR the presence of genes encoding for voltage-gated calcium channels (Cav3.1, Ca3.2, Cav3.3, Cav2.1), estrogen receptors (ESR1, ESR2, GPR30), and nuclear receptor coactivator 3 (NCOA3). Pregnancy significantly upregulated Cav3.1 and downregulated Cav3.2, Cav3.3, ESR1, ESR2, and NCOA3, compared to the non-pregnant condition. Beta-estradiol treatment (24 h, 10, 100, 1000 nM) downregulated Cav3.2, Cav3.3, Cav1.2, ESR1, ESR2, GRP30, and NCOA3 in TCs from human pregnant uterine myometrium. We also confirmed the functional expression of voltage-gated calcium channels by patch-clamp recordings and calcium imaging analysis of TCs from pregnant human myometrium by perfusing with BAY K8644, which induced calcium influx through these channels. Additionally, we demonstrated that beta-estradiol (1000 nM) antagonized the effect of BAY K8644 (2.5 or 5 µM) in the same preparations. In conclusion, we evidenced the presence of voltage-gated calcium channels and estrogen receptors in TCs from non-pregnant and pregnant human uterine myometrium and their gene expression regulation by beta-estradiol in pregnant conditions. Further exploration of the calcium signaling in TCs and its modulation by estrogen hormones will contribute to the understanding of labor and pregnancy mechanisms and to the development of effective strategies to reduce the risk of premature birth.


Subject(s)
Calcium Channels/metabolism , Estradiol/pharmacology , Myometrium/cytology , Receptors, Estrogen/metabolism , Telocytes/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Adult , Calcium Channels/genetics , Calcium Signaling/drug effects , Cells, Cultured , Down-Regulation/drug effects , Female , Humans , Middle Aged , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Telocytes/drug effects
10.
Acta Pharmacol Sin ; 39(9): 1493-1500, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29770794

ABSTRACT

Hypomotility is a common symptom of gallstone disease, which is accompanied by a loss of interstitial Cajal-like cells (ICLCs) in the gallbladder. Ursodeoxycholic acid (UDCA) is widely used in treating gallstone disease, and has shown anti-apoptotic and anti-inflammatory effects apart from its ability to dissolve gallstones. In this study, we investigated the anti-apoptotic and anti-inflammatory effects of UDCA on ICLCs in guinea pigs with gallstones. Guinea pigs were fed a high-cholesterol diet for 8 weeks to induce the formation of gallstones. A group of animals was administered UDCA (50 mg·kg-1·d-1, ig) simultaneously. At the end of 8 weeks, the animals were euthanized with anesthesia, cholecystectomy was performed immediately and gallbladder was collected for further analysis. We showed that in the model group the contractility of gallbladder muscle strips in response to both acetylcholine (ACh) and CCK-8 was severely impaired, which was significantly improved by UDCA administration. Furthermore, UDCA administration significantly reduced the apoptotic ratio of ICLCs, based on the observation of co-localization imaging of apoptotic cells and c-kit-positive cells. Western blotting analysis and real-time PCR results revealed that the TNF-α/Caspase8/Caspase3 pathway was suppressed in the UDCA-treated animals, confirming the anti-apoptotic effect of UDCA in the gallbladder. The H&E staining showed that UDCA administration significantly attenuated inflammatory cell infiltration in the gallbladder wall. In conclusion, UDCA can protect ICLCs in the gallbladder from undergoing apoptosis by inhibiting the TNF-α/Caspase8/caspase3 pathway.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis/drug effects , Protective Agents/therapeutic use , Telocytes/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Ursodeoxycholic Acid/therapeutic use , Animals , Cholagogues and Choleretics/therapeutic use , Gallbladder Emptying/drug effects , Guinea Pigs , Male , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL