1
|
Ortiz-Placín C, Salido GM, González A. Melatonin Interplay in Physiology and Disease-The Fountain of Eternal Youth Revisited. Biomolecules 2025; 15:682. [PMID: 40427575 PMCID: PMC12109172 DOI: 10.3390/biom15050682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine) is a hormone associated with the regulation of biological rhythms. The indoleamine is secreted by the pineal gland during the night, following a circadian rhythm. The highest plasmatic levels are reached during the night, whereas the lowest levels are achieved during the day. In addition to the pineal gland, other organs and tissues also produce melatonin, like, for example, the retina, Harderian glands, gut, ovaries, testes, skin, leukocytes, or bone marrow. The list of organs is extensive, including the cerebellum, airway epithelium, liver, kidney, adrenals, thymus, thyroid, pancreas, carotid body, placenta, and endometrium. At all these locations, the availability of melatonin is intended for local use. Interestingly, a decline of the circadian amplitude of the melatonin secretion occurs in old subjects in comparison to that found in younger subjects. Moreover, genetic and environmental factors are the primary causes of diseases, and oxidative stress is a key contributor to most pathologies. Numerous studies exist that show interesting effects of melatonin in different models of disease. Impairment in its secretion might have deleterious consequences for cellular physiology. In this regard, melatonin is a natural compound that is a carrier of a not yet completely known potential that deserves consideration. Thus, melatonin has emerged as a helpful ally that could be considered as a guard with powerful tools to orchestrate homeostasis in the body, majorly based on its antioxidant effects. In this review, we provide an overview of the widespread actions of melatonin against diseases preferentially affecting the elderly.
Collapse
Affiliation(s)
| | | | - Antonio González
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida de las Ciencias s/n, E-10003 Caceres, Spain; (C.O.-P.); (G.M.S.)
| |
Collapse
|
2
|
Li Q, Tang Y, Chen Y, Li B, Wang H, Liu S, Adeniran SO, Zheng P. Melatonin Regulates the Expression of VEGF and HOXA10 in Bovine Endometrial Epithelial Cells through the SIRT1/PI3K/AKT Pathway. Animals (Basel) 2024; 14:2771. [PMID: 39409719 PMCID: PMC11475476 DOI: 10.3390/ani14192771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Melatonin plays a critical role in regulating embryo attachment in ruminants. While numerous studies have investigated its effects on early embryo development in vitro, the precise mechanisms by which melatonin influences the receptivity of endometrial epithelial cells in dairy cows remain unclear. The prerequisite for embryo implantation is the specific physiological condition of the endometrium that allows the embryo to implant, also known as endometrial receptivity. In addition to this, endometrial cells undergo processes such as proliferation, differentiation, and renewal, which makes the embryo more easily implanted. In this study, bovine endometrial epithelial cells were cultured and treated with melatonin, Silent Information Regulator 1 (SIRT1) inhibitor (EX527), and protein kinase B (AKT) phosphorylation inhibitor (periposine). RT-qPCR, Western blot, and immunofluorescence analysis were performed to investigate the effects of melatonin on the expression of target gene (SIRT1); cell proliferative genes, phosphatidylinositol-4,5-bisphosphate 3-Kinase (PI3K), AKT, cyclinD1, cyclinE1; and receptive genes (Leukemia Inhibitory Factor (LIF), Vascular Endothelial Growth Factor (VEGF), Homeobox Structure Gene 10 (HOXA10)). Additionally, microRNA (miRNA) mimics and inhibitors were used to transfect the cells to study the regulatory relationship between miRNA and receptive genes. Results indicated that melatonin activates the PI3K/AKT signaling pathway, upregulates cyclinD1 and cyclinE1, and promotes the proliferation of bovine endometrial epithelial cells. Melatonin also upregulated the expression of VEGF and HOXA10 and downregulated the expression of bta-miR-497 and bta-miR-27a-3p through SIRT1/PI3K/AKT signaling pathway. Further, bta-miR-497 and bta-miR-27a-3p were found to negatively regulate VEGF and HOXA10, respectively. Therefore, melatonin regulates the expression of VEGF and HOXA10 through the SIRT1/PI3K/AKT pathway and promotes the establishment of receptivity in bovine endometrial epithelial cells.
Collapse
Affiliation(s)
- Qi Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (Q.L.); (Y.T.); (Y.C.); (B.L.); (H.W.); (S.L.)
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ying Tang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (Q.L.); (Y.T.); (Y.C.); (B.L.); (H.W.); (S.L.)
| | - Yanru Chen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (Q.L.); (Y.T.); (Y.C.); (B.L.); (H.W.); (S.L.)
| | - Bo Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (Q.L.); (Y.T.); (Y.C.); (B.L.); (H.W.); (S.L.)
| | - Hongzhan Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (Q.L.); (Y.T.); (Y.C.); (B.L.); (H.W.); (S.L.)
| | - Shicheng Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (Q.L.); (Y.T.); (Y.C.); (B.L.); (H.W.); (S.L.)
| | - Samson O. Adeniran
- Biotechnology Unit, Department of Biological Sciences, College of Basic and Applied Sciences, Mountain Top University, Ibafo 110115, Nigeria;
| | - Peng Zheng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (Q.L.); (Y.T.); (Y.C.); (B.L.); (H.W.); (S.L.)
| |
Collapse
|
3
|
Estaras M, Ortiz-Placin C, Castillejo-Rufo A, Fernandez-Bermejo M, Blanco G, Mateos JM, Vara D, Gonzalez-Cordero PL, Chamizo S, Lopez D, Rojas A, Jaen I, de Armas N, Salido GM, Iovanna JL, Santofimia-Castaño P, Gonzalez A. Melatonin controls cell proliferation and modulates mitochondrial physiology in pancreatic stellate cells. J Physiol Biochem 2023; 79:235-249. [PMID: 36334253 PMCID: PMC9905253 DOI: 10.1007/s13105-022-00930-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
We have investigated the effects of melatonin on major pathways related with cellular proliferation and energetic metabolism in pancreatic stellate cells. In the presence of melatonin (1 mM, 100 µM, 10 µM, or 1 µM), decreases in the phosphorylation of c-Jun N-terminal kinase and of p44/42 and an increase in the phosphorylation of p38 were observed. Cell viability dropped in the presence of melatonin. A rise in the phosphorylation of AMP-activated protein kinase was detected in the presence of 1 mM and 100 µM melatonin. Treatment with 1 mM melatonin decreased the phosphorylation of protein kinase B, whereas 100 µM and 10 µM melatonin increased its phosphorylation. An increase in the generation of mitochondrial reactive oxygen species and a decrease of mitochondrial membrane potential were noted following melatonin treatment. Basal and maximal respiration, ATP production by oxidative phosphorylation, spare capacity, and proton leak dropped in the presence of melatonin. The expression of complex I of the mitochondrial respiratory chain was augmented in the presence of melatonin. Conversely, in the presence of 1 mM melatonin, decreases in the expression of mitofusins 1 and 2 were detected. The glycolysis and the glycolytic capacity were diminished in cells treated with 1 mM or 100 µM melatonin. Increases in the expression of phosphofructokinase-1 and lactate dehydrogenase were noted in cells incubated with 100 µM, 10 µM, or 1 µM melatonin. The expression of glucose transporter 1 was increased in cells incubated with 10 µM or 1 µM melatonin. Conversely, 1 mM melatonin decreased the expression of all three proteins. Our results suggest that melatonin, at pharmacological concentrations, might modulate mitochondrial physiology and energy metabolism in addition to major pathways involved in pancreatic stellate cell proliferation.
Collapse
Affiliation(s)
- Matias Estaras
- Departamento de Fisiología, Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Avenida de Las Ciencias S/N, 10003, Cáceres, Spain
| | - Candido Ortiz-Placin
- Departamento de Fisiología, Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Avenida de Las Ciencias S/N, 10003, Cáceres, Spain
| | - Alba Castillejo-Rufo
- Departamento de Fisiología, Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Avenida de Las Ciencias S/N, 10003, Cáceres, Spain
| | | | - Gerardo Blanco
- Unidad de Cirugía Hepatobiliopancreática Y Transplante Hepático, Hospital Universitario, Badajoz, Spain
| | - Jose M Mateos
- Departamento de Gastroenterología, Hospital Universitario, Cáceres, Spain
| | - Daniel Vara
- Departamento de Gastroenterología, Hospital Universitario, Cáceres, Spain
| | | | - Sandra Chamizo
- Departamento de Gastroenterología, Hospital Universitario, Cáceres, Spain
| | - Diego Lopez
- Unidad de Cirugía Hepatobiliopancreática Y Transplante Hepático, Hospital Universitario, Badajoz, Spain
| | - Adela Rojas
- Unidad de Cirugía Hepatobiliopancreática Y Transplante Hepático, Hospital Universitario, Badajoz, Spain
| | - Isabel Jaen
- Unidad de Cirugía Hepatobiliopancreática Y Transplante Hepático, Hospital Universitario, Badajoz, Spain
| | - Noelia de Armas
- Unidad de Cirugía Hepatobiliopancreática Y Transplante Hepático, Hospital Universitario, Badajoz, Spain
| | - Gines M Salido
- Departamento de Fisiología, Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Avenida de Las Ciencias S/N, 10003, Cáceres, Spain
| | - Juan L Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique Et Technologique de Luminy, Marseille, France
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique Et Technologique de Luminy, Marseille, France
| | - Antonio Gonzalez
- Departamento de Fisiología, Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Avenida de Las Ciencias S/N, 10003, Cáceres, Spain.
| |
Collapse
|
4
|
Estaras M, Martinez R, Garcia A, Ortiz-Placin C, Iovanna JL, Santofimia-Castaño P, Gonzalez A. Melatonin modulates metabolic adaptation of pancreatic stellate cells subjected to hypoxia. Biochem Pharmacol 2022; 202:115118. [PMID: 35671789 DOI: 10.1016/j.bcp.2022.115118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022]
Abstract
Pancreatic stellate cells (PSCs), the main cell type responsible for the development of fibrosis in pancreatic cancer, proliferate actively under hypoxia. Melatonin has received attention as a potential antifibrotic agent due to its anti-proliferative actions on PSCs. In this work, we investigated the activation of the PI3K/Akt/mTOR pathway and the metabolic adaptations that PSCs undergo under hypoxic conditions, as well as the probable modulation by melatonin. Incubation of cells under hypoxia induced an increase in cell proliferation, and in the expression of alpha-smooth muscle actin and of collagen type 1. In addition, an increase in the phosphorylation of Akt was observed, whereas a decrease in the phosphorylation of PTEN and GSK-3b was noted. The phosphorylation of mTOR and its substrate p70 S6K was decreased under hypoxia. Treatment of PSCs with melatonin under hypoxia diminished cell proliferation, the levels of alpha-smooth muscle actin and of collagen type 1, the phosphorylation of Akt and increased phosphorylation of mTOR. Mitochondrial activity decreased in PSCs under hypoxia. A glycolytic shift was observed. Melatonin further decreased mitochondrial activity. Under hypoxia, no increase in autophagic flux was noted. However, melatonin treatment induced autophagy activation. Nevertheless, inhibition of this process did not induce detectable changes in the viability of cells treated with melatonin. We conclude that PSCs undergo metabolic adaptation under hypoxia that might help them survive and that pharmacological concentrations of melatonin modulate cell responses to hypoxia. Our results contribute to the knowledge of the mechanisms by which melatonin could modulate fibrosis within the pancreas.
Collapse
Affiliation(s)
- Matias Estaras
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, Cáceres, España
| | - Remigio Martinez
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de Extremadura, Cáceres, España
| | - Alfredo Garcia
- Departamento de Producción Animal, CICYTEX-La Orden, Guadajira, Badajoz, España
| | - Candido Ortiz-Placin
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, Cáceres, España
| | - Juan L Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Antonio Gonzalez
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, Cáceres, España.
| |
Collapse
|
5
|
Fenton-Navarro B, Garduño Ríos D, Torner L, Letechipía-Vallejo G, Cervantes M. Melatonin Decreases Circulating Levels of Galectin-3 and Cytokines, Motor Activity, and Anxiety Following Acute Global Cerebral Ischemia in Male Rats. Arch Med Res 2021; 52:505-513. [DOI: 10.1016/j.arcmed.2021.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/21/2020] [Accepted: 01/21/2021] [Indexed: 12/23/2022]
|
6
|
Estaras M, Gonzalez-Portillo MR, Fernandez-Bermejo M, Mateos JM, Vara D, Blanco-Fernandez G, Lopez-Guerra D, Roncero V, Salido GM, González A. Melatonin Induces Apoptosis and Modulates Cyclin Expression and MAPK Phosphorylation in Pancreatic Stellate Cells Subjected to Hypoxia. Int J Mol Sci 2021; 22:5555. [PMID: 34074034 PMCID: PMC8197391 DOI: 10.3390/ijms22115555] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023] Open
Abstract
In certain diseases of the pancreas, pancreatic stellate cells form an important part of fibrosis and are critical for the development of cancer cells. A hypoxic condition develops within the tumor, to which pancreatic stellate cells adapt and are able to proliferate. The consequence is the growth of the tumor. Melatonin, the product of the pineal gland, is gaining attention as an agent with therapeutic potential against pancreatic cancers. Its actions on tumor cells lead, in general, to a reduction in cell viability and proliferation. However, its effects on pancreatic stellate cells subjected to hypoxia are less known. In this study, we evaluated the actions of pharmacological concentrations of melatonin (1 mM-1 µM) on pancreatic stellate cells subjected to hypoxia. The results show that melatonin induced a decrease in cell viability at the highest concentrations tested. Similarly, the incorporation of BrdU into DNA was diminished by melatonin. The expression of cyclins A and D also was decreased in the presence of melatonin. Upon treatment of cells with melatonin, increases in the expression of major markers of ER stress, namely BIP, phospho-eIF2α and ATF-4, were detected. Modulation of apoptosis was noticed as an increase in caspase-3 activation. In addition, changes in the phosphorylated state of p44/42, p38 and JNK MAPKs were detected in cells treated with melatonin. A slight decrease in the content of α-smooth muscle actin was detected in cells treated with melatonin. Finally, treatment of cells with melatonin decreased the expression of matrix metalloproteinases 2, 3, 9 and 13. Our observations suggest that melatonin, at pharmacological concentrations, diminishes the proliferation of pancreatic stellate cells subjected to hypoxia through modulation of cell cycle, apoptosis and the activation of crucial MAPKs. Cellular responses might involve certain ER stress regulator proteins. In view of the results, melatonin could be taken into consideration as a potential therapeutic agent for pancreatic fibrosis.
Collapse
Affiliation(s)
- Matias Estaras
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| | - Manuel R. Gonzalez-Portillo
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| | - Miguel Fernandez-Bermejo
- Department of Gastroenterology, San Pedro de Alcantara Hospital, 10003 Caceres, Spain; (M.F.-B.); (J.M.M.); (D.V.)
| | - Jose M. Mateos
- Department of Gastroenterology, San Pedro de Alcantara Hospital, 10003 Caceres, Spain; (M.F.-B.); (J.M.M.); (D.V.)
| | - Daniel Vara
- Department of Gastroenterology, San Pedro de Alcantara Hospital, 10003 Caceres, Spain; (M.F.-B.); (J.M.M.); (D.V.)
| | - Gerardo Blanco-Fernandez
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, University Hospital, 06080 Badajoz, Spain; (G.B.-F.); (D.L.-G.)
| | - Diego Lopez-Guerra
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, University Hospital, 06080 Badajoz, Spain; (G.B.-F.); (D.L.-G.)
| | - Vicente Roncero
- Unit of Histology and Pathological Anatomy, Veterinary Faculty, University of Extremadura, 10003 Caceres, Spain;
| | - Gines M. Salido
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| | - Antonio González
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| |
Collapse
|
7
|
Estaras M, Gonzalez-Portillo MR, Martinez R, Garcia A, Estevez M, Fernandez-Bermejo M, Mateos JM, Vara D, Blanco-Fernández G, Lopez-Guerra D, Roncero V, Salido GM, Gonzalez A. Melatonin Modulates the Antioxidant Defenses and the Expression of Proinflammatory Mediators in Pancreatic Stellate Cells Subjected to Hypoxia. Antioxidants (Basel) 2021; 10:577. [PMID: 33918063 PMCID: PMC8070371 DOI: 10.3390/antiox10040577] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic stellate cells (PSC) play a major role in the formation of fibrotic tissue in pancreatic tumors. On its side, melatonin is a putative therapeutic agent for pancreatic cancer and inflammation. In this work, the actions of melatonin on PSC subjected to hypoxia were evaluated. Reactive oxygen species (ROS) generation reduced (GSH) and oxidized (GSSG) levels of glutathione, and protein and lipid oxidation were analyzed. The phosphorylation of nuclear factor erythroid 2-related factor (Nrf2), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), and the regulatory protein nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor-alpha (IκBα) was studied. The expression of Nrf2-regulated antioxidant enzymes, superoxide dismutase (SOD) enzymes, cyclooxygenase 2 (COX-2), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were also studied. Total antioxidant capacity (TAC) was assayed. Finally, cell viability was studied. Under hypoxia and in the presence of melatonin generation of ROS was observed. No increases in the oxidation of proteins or lipids were detected. The phosphorylation of Nrf2 and the expression of the antioxidant enzymes catalytic subunit of glutamate-cysteine ligase, catalase, NAD(P)H-quinone oxidoreductase 1, heme oxygenase-1, SOD1, and of SOD2 were augmented. The TAC was increased. Protein kinase C was involved in the effects of melatonin. Melatonin decreased the GSH/GSSG ratio at the highest concentration tested. Cell viability dropped in the presence of melatonin. Finally, melatonin diminished the phosphorylation of NF-kB and the expression of COX-2, IL-6, and TNF-α. Our results indicate that melatonin, at pharmacological concentrations, modulates the red-ox state, viability, and the expression of proinflammatory mediators in PSC subjected to hypoxia.
Collapse
Affiliation(s)
- Matias Estaras
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| | - Manuel R. Gonzalez-Portillo
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| | - Remigio Martinez
- Department of Animal Health, Veterinary Faculty, University of Extremadura, 10003 Caceres, Spain;
| | - Alfredo Garcia
- Department of Animal Production, CICYTEX-La Orden, 06187 Badajoz, Spain;
| | - Mario Estevez
- IPROCAR Research Institute, Food Technology, University of Extremadura, 10003 Cáceres, Spain;
| | - Miguel Fernandez-Bermejo
- Department of Gastroenterology, San Pedro de Alcantara Hospital, 10003 Caceres, Spain; (M.F.-B.); (J.M.M.); (D.V.)
| | - Jose M. Mateos
- Department of Gastroenterology, San Pedro de Alcantara Hospital, 10003 Caceres, Spain; (M.F.-B.); (J.M.M.); (D.V.)
| | - Daniel Vara
- Department of Gastroenterology, San Pedro de Alcantara Hospital, 10003 Caceres, Spain; (M.F.-B.); (J.M.M.); (D.V.)
| | - Gerardo Blanco-Fernández
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, Infanta Cristina Hospital, 06080 Badajoz, Spain; (G.B.-F.); (D.L.-G.)
| | - Diego Lopez-Guerra
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, Infanta Cristina Hospital, 06080 Badajoz, Spain; (G.B.-F.); (D.L.-G.)
| | - Vicente Roncero
- Unit of Histology and Pathological Anatomy, Veterinary Faculty, University of Extremadura, 10003 Caceres, Spain;
| | - Gines M. Salido
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| | - Antonio Gonzalez
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| |
Collapse
|
8
|
Estaras M, Marchena AM, Fernandez-Bermejo M, Mateos JM, Vara D, Roncero V, Salido GM, Gonzalez A. The melatonin receptor antagonist luzindole induces the activation of cellular stress responses and decreases viability of rat pancreatic stellate cells. J Appl Toxicol 2020; 40:1554-1565. [PMID: 32567733 DOI: 10.1002/jat.4018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 12/22/2022]
Abstract
In this study, we have examined the effects of luzindole, a melatonin receptor-antagonist, on cultured pancreatic stellate cells. Intracellular free-Ca2+ concentration, production of reactive oxygen species (ROS), activation of mitogen-activated protein kinases (MAPK), endoplasmic reticulum stress and cell viability were analyzed. Stimulation of cells with the luzindole (1, 5, 10 and 50 μm) evoked a slow and progressive increase in intracellular free Ca2+ ([Ca2+ ]i ) towards a plateau. The effect of the compound on Ca2+ mobilization depended on the concentration used. Incubation of cells with the sarcoendoplasmic reticulum Ca2+ -ATPase inhibitor thapsigargin (1 μm), in the absence of Ca2+ in the extracellular medium, induced a transient increase in [Ca2+ ]i . In the presence of thapsigargin, the addition of luzindole to the cells failed to induce further mobilization of Ca2+ . Luzindole induced a concentration-dependent increase in ROS generation, both in the cytosol and in the mitochondria. This effect was smaller in the absence of extracellular Ca2+ . In the presence of luzindole the phosphorylation of p44/42 and p38 MAPKs was increased, whereas no changes in the phosphorylation of JNK could be noted. Moreover, the detection of the endoplasmic reticulum stress-sensor BiP was increased in the presence of luzindole. Finally, viability was decreased in cells treated with luzindole. Because cellular membrane receptors for melatonin have not been detected in pancreatic stellate cells, we conclude that luzindole could exert direct effects that are not mediated through its action on melatonin membrane receptors.
Collapse
Affiliation(s)
- Matias Estaras
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| | - Ana M Marchena
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| | | | - Jose M Mateos
- Department of Gastroenterology, San Pedro de Alcantara Hospital, Caceres, Spain
| | - Daniel Vara
- Department of Gastroenterology, San Pedro de Alcantara Hospital, Caceres, Spain
| | - Vicente Roncero
- Unit of Histology and Pathological Anatomy, Veterinary Faculty, University of Extremadura, Caceres, Spain
| | - Gines M Salido
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| | - Antonio Gonzalez
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| |
Collapse
|
9
|
Estaras M, Martinez-Morcillo S, García A, Martinez R, Estevez M, Perez-Lopez M, Miguez MP, Fernandez-Bermejo M, Mateos JM, Vara D, Blanco G, Lopez D, Roncero V, Salido GM, Gonzalez A. Pancreatic stellate cells exhibit adaptation to oxidative stress evoked by hypoxia. Biol Cell 2020; 112:280-299. [PMID: 32632968 DOI: 10.1111/boc.202000020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/31/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND INFORMATION Pancreatic stellate cells play a key role in the fibrosis that develops in diseases such as pancreatic cancer. In the growing tumour, a hypoxia condition develops under which cancer cells are able to proliferate. The growth of fibrotic tissue contributes to hypoxia. In this study, the effect of hypoxia (1% O2 ) on pancreatic stellate cells physiology was investigated. Changes in intracellular free-Ca2+ concentration, mitochondrial free-Ca2+ concentration and mitochondrial membrane potential were studied by fluorescence techniques. The status of enzymes responsible for the cellular oxidative state was analyzed by quantitative reverse transcription-polymerase chain reaction, high-performance liquid chromatography, spectrophotometric and fluorimetric methods and by Western blotting analysis. Cell viability and proliferation were studied by crystal violet test, 5-bromo-2-deoxyuridine cell proliferation test and Western blotting analysis. Finally, cell migration was studied employing the wound healing assay. RESULTS Hypoxia induced an increase in intracellular and mitochondrial free-Ca2+ concentration, whereas mitochondrial membrane potential was decreased. An increase in mitochondrial reactive oxygen species production was observed. Additionally, an increase in the oxidation of proteins and lipids was detected. Moreover, cellular total antioxidant capacity was decreased. Increases in the expression of superoxide dismutase 1 and 2 were observed and superoxide dismutase activity was augmented. Hypoxia evoked a decrease in the oxidized/reduced glutathione ratio. An increase in the phosphorylation of nuclear factor erythroid 2-related factor and in expression of the antioxidant enzymes catalytic subunit of glutamate-cysteine ligase, catalase, NAD(P)H-quinone oxidoreductase 1 and heme oxygenase-1 were detected. The expression of cyclin A was decreased, whereas expression of cyclin D and the content of 5-bromo-2-deoxyuridine were increased. This was accompanied by an increase in cell viability. The phosphorylation state of c-Jun NH2 -terminal kinase was increased, whereas that of p44/42 and p38 was decreased. Finally, cells subjected to hypoxia maintained migration ability. CONCLUSIONS AND SIGNIFICANCE Hypoxia creates pro-oxidant conditions in pancreatic stellate cells to which cells adapt and leads to increased viability and proliferation.
Collapse
Affiliation(s)
- Matias Estaras
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| | | | - Alfredo García
- Department of Animal Production, Cicytex-La Orden, Badajoz, Spain
| | - Remigio Martinez
- Department of Animal Health, Veterinary Faculty, University of Extremadura, Caceres, Spain
| | - Mario Estevez
- IPROCAR Research Institute, Food Technology, University of Extremadura, Caceres, 10003, Spain
| | - Marcos Perez-Lopez
- Unit of Toxicology, Veterinary Faculty, University of Extremadura, Caceres, Spain
| | - Maria P Miguez
- Unit of Toxicology, Veterinary Faculty, University of Extremadura, Caceres, Spain
| | | | - Jose M Mateos
- Department of Gastroenterology, San Pedro de Alcantara Hospital, Caceres, Spain
| | - Daniel Vara
- Department of Gastroenterology, San Pedro de Alcantara Hospital, Caceres, Spain
| | - Gerardo Blanco
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, Infanta Cristina Hospital, Badajoz, Spain
| | - Diego Lopez
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, Infanta Cristina Hospital, Badajoz, Spain
| | - Vicente Roncero
- Unit of Histology and Pathological Anatomy, Veterinary Faculty, University of Extremadura, Caceres, Spain
| | - Gines M Salido
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| | - Antonio Gonzalez
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| |
Collapse
|
10
|
Radoslavova S, Ouadid-Ahidouch H, Prevarskaya N. Ca2+ signaling is critical for pancreatic stellate cell’s pathophysiology : from fibrosis to cancer hallmarks. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
11
|
Tiong YL, Ng KY, Koh RY, Ponnudurai G, Chye SM. Melatonin promotes Schwann cell dedifferentiation and proliferation through the Ras/Raf/ERK and MAPK pathways, and glial cell-derived neurotrophic factor expression. Exp Ther Med 2020; 20:16. [PMID: 32934681 PMCID: PMC7471953 DOI: 10.3892/etm.2020.9143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/22/2019] [Indexed: 12/24/2022] Open
Abstract
Upon peripheral nerve injury (PNI), continuous proliferation of Schwann cells is critical for axon regeneration and tubular reconstruction for nerve regeneration. Melatonin is a hormone that is able to induce proliferation in various cell types. In the present study, the effects of melatonin on promoting Schwann cell proliferation and the molecular mechanism involved were investigated. The present results showed that melatonin enhanced the melatonin receptors (MT1 and MT2) expression in Schwann cells. Melatonin induced Schwann cell dedifferentiation into progenitor-like Schwann cells, as observed by immunofluorescence staining, which showed Sox2 marker expression. In addition, melatonin enhanced Schwann cell proliferation, mediated by the upregulation of glial cell-derived neurotropic factor (GNDF) and protein kinase C (PKC). Furthermore, the Ras/Raf/ERK and MAPK signaling pathways were also involved in Schwann cell dedifferentiation and proliferation. In conclusion, melatonin induced Schwann cell dedifferentiation and proliferation via the Ras/Raf/ERK, MAPK and GDNF/PKC pathways. The present results suggested that melatonin could be used to enhance the recovery of PNI.
Collapse
Affiliation(s)
- Yee Lian Tiong
- School of Postgraduate, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor 47500, Malaysia
| | - Rhun Yian Koh
- School of Health Science, International Medical University, Kuala Lumpur 57000, Malaysia
| | | | - Soi Moi Chye
- School of Health Science, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
12
|
Bae H, Yang C, Lee JY, Park S, Bazer FW, Song G, Lim W. Melatonin improves uterine-conceptus interaction via regulation of SIRT1 during early pregnancy. J Pineal Res 2020; 69:e12670. [PMID: 32421880 DOI: 10.1111/jpi.12670] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
Melatonin has been shown to improve in vitro fertilization and offspring survival after bacterial infection, but its role in regulating maternal-fetal communication during early pregnancy has not been investigated. Results of this study demonstrated expression of abundant melatonin receptors in conceptus and endometrium during early pregnancy. In gilts, expression of melatonin receptor 1A (MTNR1A or MT1) and melatonin receptor 1B (MTNR1B or MT2) increased in trophectoderm (Tr) and uterine luminal epithelium (LE) with advancing days during early pregnancy in a different manner. Melatonin increased proliferation and migration of porcine trophectoderm (pTr) cell, the percent pTr cells in the G2 phase of the cell cycle, and the expression of implantation-related genes by pTr cells and endometrial luminal epithelium (pLE). Melatonin also attenuated the production of LPS-induced pro-inflammatory cytokines and tunicamycin-induced endoplasmic reticulum (ER) stress-sensing proteins. The expression of sirtuin 1 (SIRT1) as a potential target of melatonin increased between Days 9 and 14 of gestation. Co-treatment with SIRT1 inhibitor EX527 and melatonin restored cell-cell interactions through PI3K and MAPK signaling. Knockdown of SIRT1 decreased the expression of implantation-related genes, as well as migration of pTr and pLE cells. The expression of microRNAs regulated by SIRT1 was suppressed in response to melatonin. Furthermore, melatonin significantly increased lipopolysaccharide (LPS)-reduced fertilization and embryogenesis in zebrafish model. These results suggest that melatonin may improve the uterine-conceptus interactions via the regulation of SIRT1 during early pregnancy.
Collapse
Affiliation(s)
- Hyocheol Bae
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Changwon Yang
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Jin-Young Lee
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sunwoo Park
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Fuller W Bazer
- Department of Animal Science, Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, TX, USA
| | - Gwonhwa Song
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Whasun Lim
- Department of Food and Nutrition, College of Science and Technology, Kookmin University, Seoul, Korea
| |
Collapse
|
13
|
Estaras M, Peña FJ, Tapia JA, Fernandez-Bermejo M, Mateos JM, Vara D, Roncero V, Blanco G, Lopez D, Salido GM, Gonzalez A. Melatonin modulates proliferation of pancreatic stellate cells through caspase-3 activation and changes in cyclin A and D expression. J Physiol Biochem 2020; 76:345-355. [PMID: 32361979 DOI: 10.1007/s13105-020-00740-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022]
Abstract
In this study, the effects of melatonin (1 μM-1 mM) on pancreatic stellate cells (PSC) have been examined. Cell viability and proliferation, caspase-3 activation, and the expression of cyclin A and cyclin D were analyzed. Our results show that melatonin decreased PSC viability in a time- and concentration-dependent manner. This effect was not inhibited by treatment of cells with MT1, MT2, calmodulin, or ROR-alpha inhibitors prior to melatonin addition. Activation of caspase-3 in response to melatonin was detected. The expression of cyclin A and cyclin D was decreased in cells treated with melatonin. Finally, changes in BrdU incorporation into the newly synthesized DNA of proliferating cells were also observed in the presence of melatonin. We conclude that melatonin, at pharmacological concentrations, modulates proliferation of PSC through activation of apoptosis and involving crucial regulators of the cell cycle. These actions might not require specific melatonin receptors. Our observations suggest that melatonin, at high doses, could potentially exert anti-fibrotic effects and, thus, could be taken into consideration as supportive treatment in the therapy of pancreatic diseases.
Collapse
Affiliation(s)
- Matias Estaras
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida de las Ciencias s/n, E-10003, Cáceres, Spain
| | - Fernando J Peña
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - José A Tapia
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida de las Ciencias s/n, E-10003, Cáceres, Spain
| | | | - Jose M Mateos
- Department of Gastroenterology, San Pedro de Alcantara Hospital, Cáceres, Spain
| | - Daniel Vara
- Department of Gastroenterology, San Pedro de Alcantara Hospital, Cáceres, Spain
| | - Vicente Roncero
- Unit of Histology and Pathological Anatomy, Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | - Gerardo Blanco
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, Infanta Cristina Hospital, Badajoz, Spain
| | - Diego Lopez
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, Infanta Cristina Hospital, Badajoz, Spain
| | - Gines M Salido
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida de las Ciencias s/n, E-10003, Cáceres, Spain
| | - Antonio Gonzalez
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida de las Ciencias s/n, E-10003, Cáceres, Spain.
| |
Collapse
|
14
|
Gonzalez A, Estaras M, Martinez-Morcillo S, Martinez R, García A, Estévez M, Santofimia-Castaño P, Tapia JA, Moreno N, Pérez-López M, Míguez MP, Blanco-Fernández G, Lopez-Guerra D, Fernandez-Bermejo M, Mateos JM, Vara D, Roncero V, Salido GM. Melatonin modulates red-ox state and decreases viability of rat pancreatic stellate cells. Sci Rep 2020; 10:6352. [PMID: 32286500 PMCID: PMC7156707 DOI: 10.1038/s41598-020-63433-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
In this work we have studied the effects of pharmacological concentrations of melatonin (1 µM-1 mM) on pancreatic stellate cells (PSC). Cell viability was analyzed by AlamarBlue test. Production of reactive oxygen species (ROS) was monitored following CM-H2DCFDA and MitoSOX Red-derived fluorescence. Total protein carbonyls and lipid peroxidation were analyzed by HPLC and spectrophotometric methods respectively. Mitochondrial membrane potential (ψm) was monitored by TMRM-derived fluorescence. Reduced (GSH) and oxidized (GSSG) levels of glutathione were determined by fluorescence techniques. Quantitative reverse transcription-polymerase chain reaction was employed to detect the expression of Nrf2-regulated antioxidant enzymes. Determination of SOD activity and total antioxidant capacity (TAC) were carried out by colorimetric methods, whereas expression of SOD was analyzed by Western blotting and RT-qPCR. The results show that melatonin decreased PSC viability in a concentration-dependent manner. Melatonin evoked a concentration-dependent increase in ROS production in the mitochondria and in the cytosol. Oxidation of proteins was detected in the presence of melatonin, whereas lipids oxidation was not observed. Depolarization of ψm was noted with 1 mM melatonin. A decrease in the GSH/GSSG ratio was observed, that depended on the concentration of melatonin used. A concentration-dependent increase in the expression of the antioxidant enzymes catalytic subunit of glutamate-cysteine ligase, catalase, NAD(P)H-quinone oxidoreductase 1 and heme oxygenase-1 was detected in cells incubated with melatonin. Finally, decreases in the expression and in the activity of superoxide dismutase were observed. We conclude that pharmacological concentrations melatonin modify the redox state of PSC, which might decrease cellular viability.
Collapse
Affiliation(s)
- Antonio Gonzalez
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain.
| | - Matias Estaras
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| | | | - Remigio Martinez
- Department of Animal Health, Veterinary Faculty, University of Extremadura, Caceres, Spain
| | - Alfredo García
- Department of Animal Production, CICYTEX-La Orden, Guadajira, Badajoz, Spain
| | - Mario Estévez
- IPROCAR Research Institute, Food Technology, University of Extremadura, 10003, Cáceres, Spain
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Jose A Tapia
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| | - Noelia Moreno
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| | - Marcos Pérez-López
- Unit of Toxicology, Veterinary Faculty, University of Extremadura, Caceres, Spain
| | - María P Míguez
- Unit of Toxicology, Veterinary Faculty, University of Extremadura, Caceres, Spain
| | - Gerardo Blanco-Fernández
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, Infanta Cristina Hospital, Badajoz, Spain
| | - Diego Lopez-Guerra
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, Infanta Cristina Hospital, Badajoz, Spain
| | | | - Jose M Mateos
- Department of Gastroenterology, San Pedro de Alcantara Hospital, Caceres, Spain
| | - Daniel Vara
- Department of Gastroenterology, San Pedro de Alcantara Hospital, Caceres, Spain
| | - Vicente Roncero
- Unit of Histology and Pathological Anatomy, Veterinary Faculty, University of Extremadura, Caceres, Spain
| | - Gines M Salido
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| |
Collapse
|
15
|
Bertolini M, Ramot Y, Gherardini J, Heinen G, Chéret J, Welss T, Giesen M, Funk W, Paus R. Theophylline exerts complex anti-ageing and anti-cytotoxicity effects in human skin ex vivo. Int J Cosmet Sci 2019; 42:79-88. [PMID: 31633195 DOI: 10.1111/ics.12589] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/17/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Theophylline is a phosphodiesterase inhibitor that is being used clinically for asthma therapy. In addition, it is recognized as a cosmetic agent with possible anti-ageing and anti-oxidative properties. Nevertheless, how it affects human skin is still poorly examined. METHODS Theophylline (10 or 100 µM) was administered to the culture medium of full-thickness human skin ex vivo for 24 or 72 h. RESULTS Theophylline stimulated protein expression of the anti-oxidant metallothionein-1 and mRNA levels of collagen I and III. Assessment of fibrillin-1 immunohistology revealed enhanced structural stability of dermal microfibrils. Theophylline also exerted extracellular matrix-protective effects by decreasing MMP-2 and MMP-9 mRNA levels, partially antagonizing the effects of menadione, the potent, toxic ROS donor. In addition, it decreased menadione-stimulated epidermal keratinocytes apoptosis. Interestingly, theophylline also increased the level of intracutaneously produced melatonin, that is the most potent ROS-protective and DNA damage repair neuromediator, and tendentially increased protein expression of MT1, the melatonin receptor. Theophylline also increased the expression of keratin 15, the stem cell marker, in the epidermal basal layer but did not change mitochondrial activity or epidermal pigmentation. CONCLUSION This ex vivo pilot study in human skin shows that theophylline possesses several interesting complex skin-protective properties. It encourages further examination of theophylline as a topical candidate for anti-ageing treatment.
Collapse
Affiliation(s)
- M Bertolini
- Monasterium Laboratory GmbH, 48149 , Muenster, Germany
| | - Y Ramot
- Department of Dermatology, The Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, 9112001 , Jerusalem, Israel
| | - J Gherardini
- Monasterium Laboratory GmbH, 48149 , Muenster, Germany
| | - G Heinen
- Henkel AG & Co. KGaA, 40589 , Düsseldorf, Germany
| | - J Chéret
- Monasterium Laboratory GmbH, 48149 , Muenster, Germany.,Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 33136 , Miami, FL, USA
| | - T Welss
- Henkel AG & Co. KGaA, 40589 , Düsseldorf, Germany
| | - M Giesen
- Henkel AG & Co. KGaA, 40589 , Düsseldorf, Germany
| | - W Funk
- Clinic for Plastic, Aesthetic and Reconstructive Surgery, Dr. Dr. med. Funk, 81739, Munich, Germany
| | - R Paus
- Monasterium Laboratory GmbH, 48149 , Muenster, Germany.,Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 33136 , Miami, FL, USA.,Centre for Dermatology Research, University of Manchester, M13 9PL, Manchester, UK
| |
Collapse
|
16
|
Huang KF, Ma KH, Jhap TY, Liu PS, Chueh SH. Ultraviolet B irradiation induced Nrf2 degradation occurs via activation of TRPV1 channels in human dermal fibroblasts. Free Radic Biol Med 2019; 141:220-232. [PMID: 31220549 DOI: 10.1016/j.freeradbiomed.2019.06.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 06/10/2019] [Accepted: 06/15/2019] [Indexed: 12/21/2022]
Abstract
Ultraviolet (UV) irradiation causes cellular oxidative stress. Under redox imbalance, Keap1-dependent Nrf2 degradation is minimal. In this study, we examined the role of Ca2+ in Nrf2 homeostasis after UVB irradiation using human dermal fibroblasts. UVB irradiation stimulates 12-lipoxygenase and the product 12-hydroxyeicosatetraenoic acid then activates TRPV1 increasing the cell's cytosolic Ca2+ concentration. UVB irradiation induced reactive oxygen species generation and apoptosis are inhibited in the absence of Ca2+ or in the presence of either a 12-lipoxygenase inhibitor or a TRPV1 inhibitor during and after UVB irradiation. Thus, the Ca2+ increase via TRPV1 is a critical factor in UVB irradiation induced oxidative stress. UVB irradiation induces a Ca2+ dependent Nrf2 degradation and thus activation of TRPV1 with 12-hydroxyeicosatetraenoic acid also decreasing Nrf2 levels. UVB irradiation induced Nrf2 degradation is inhibited by co-treatment of cells with W-7, cyclosporin A, SB-216763 or MG-132, which are inhibitors of calmodulin, calcineurin, GSK3β and the proteasome, respectively. Furthermore, UVB irradiation in parallel induces GSK3β dephosphorylation in a Ca2+ dependent manner. Co-immunoprecipitation showed that UVB irradiation induces an increase in Nrf2 phosphorylation, an increase in the binding of β-TrCP and Nrf2, and an increase in Nrf2 ubiquitination; these effects are all Ca2+ dependent. These findings suggest that UVB irradiation induced GSK3β activation in a Ca2+ dependent manner, which then stimulates the phosphorylation and ubiquitination of Nrf2 via β-TrCP. Indeed, silencing of β-TrCP was found to inhibit UVB irradiation-induced oxidative stress, Nrf2 degradation and apoptosis, while it had no effect on the Ca2+ increase. Taken together, our results suggest that a Ca2+ influx via TRPV1 is responsible for UVB irradiation-induced Nrf2 degradation and that modulation of the Ca2+-calmodulin-calcineurin-GSK3β-Nrf2-β-TrCP-Cullin-1 pathway may explain Ca2+ dependent Nrf2 degradation.
Collapse
Affiliation(s)
- Kuo-Feng Huang
- Division of Plastic Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan, ROC
| | - Kuo-Hsing Ma
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Tian-You Jhap
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Pei-Shan Liu
- Department of Microbiology, Soochow University, Taipei, Taiwan, ROC
| | - Sheau-Huei Chueh
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
17
|
Estaras M, Moreno N, Santofimia-Castaño P, Martinez-Morcillo S, Roncero V, Blanco G, Lopez D, Fernandez-Bermejo M, Mateos JM, Iovanna JL, Salido GM, Gonzalez A. Melatonin induces reactive oxygen species generation and changes in glutathione levels and reduces viability in human pancreatic stellate cells. J Physiol Biochem 2019; 75:185-197. [PMID: 30868511 DOI: 10.1007/s13105-019-00671-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/26/2019] [Indexed: 12/12/2022]
Abstract
In this study, the effects of pharmacological concentrations of melatonin (1 μM-1 mM) on human pancreatic stellate cells (HPSCs) have been examined. Cell type-specific markers and expression of melatonin receptors were analyzed by western blot analysis. Changes in intracellular free Ca2+ concentration were followed by fluorimetric analysis of fura-2-loaded cells. Reduced glutathione (GSH) and oxidized glutathione (GSSG) levels were determined by fluorescence techniques. Production of reactive oxygen species (ROS) was monitored following 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate acetyl ester and MitoSOX™ Red-derived fluorescence. Cell viability was studied using the AlamarBlue® test. Cultured cells expressed markers typical of stellate cells. However, cell membrane receptors for melatonin could not be detected. Thapsigargin, bradykinin, or melatonin induced changes in intracellular free Ca2+ concentration. In the presence of the indole, a decrease in the GSH/GSSG ratio was observed that depended on the concentration of melatonin used. Furthermore, the indole evoked a concentration-dependent increase in ROS production in the mitochondria and in the cytosol. Finally, melatonin decreased HPSC viability in a time and concentration-dependent manner. We conclude that melatonin, at pharmacological concentrations, induces changes in the oxidative state of HPSC. This might regulate cellular viability and could not involve specific plasma membrane receptors.
Collapse
Affiliation(s)
- Matias Estaras
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida Universidad s/n, 10003, Cáceres, Spain
| | - Noelia Moreno
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida Universidad s/n, 10003, Cáceres, Spain
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | | | - Vicente Roncero
- Unit of Histology and Pathological Anatomy, Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | - Gerardo Blanco
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, Infanta Cristina Hospital, Badajoz, Spain
| | - Diego Lopez
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, Infanta Cristina Hospital, Badajoz, Spain
| | | | - Jose M Mateos
- Department of Gastroenterology, San Pedro de Alcantara Hospital, Cáceres, Spain
| | - Juan L Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Gines M Salido
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida Universidad s/n, 10003, Cáceres, Spain
| | - Antonio Gonzalez
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida Universidad s/n, 10003, Cáceres, Spain.
| |
Collapse
|
18
|
Martínez-Morcillo S, Pérez-López M, Soler-Rodríguez F, González A. The organophosphorus pesticide dimethoate decreases cell viability and induces changes in different biochemical parameters of rat pancreatic stellate cells. Toxicol In Vitro 2019; 54:89-97. [PMID: 30243730 DOI: 10.1016/j.tiv.2018.09.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 09/15/2018] [Accepted: 09/18/2018] [Indexed: 12/14/2022]
Abstract
In the present study we employed cultured pancreatic stellate cells to study the effect of the organophosphorus insecticide dimethoate on pancreatic cell physiology. Esterase activity, cell viability, reactive oxygen species generation and Ca2+ mobilization were examined. Our results show that dimethoate (0.1, 1 and 10 μM) induced a concentration-dependent inhibition of cholinesterase enzymatic activity at all concentrations tested. A drop in carboxylesterase activity was noted in the presence of 10 μM dimethoate. In the presence of the pesticide a decrease in cell viability was detected. The clearer effect could be observed when the cells had been incubated during 96 h in the presence of dimethoate. The pesticide induced a slight but statistically significant increase in the production of reactive oxygen species in the mitochondria. Incubation of cells with dimethoate, in the presence of Ca2+ in the extracellular medium, led to a slow and progressive increase in [Ca2+]c towards an elevated value over the prestimulation level. A similar behavior was observed in the absence of extracellular Ca2+, indicating that dimethoate releases Ca2+ from the intracellular stores. Our results suggest that dimethoate might alter intracellular pathways that are critical for pancreatic physiology, creating a situation potentially leading to dysfunction in the exocrine pancreas.
Collapse
Affiliation(s)
| | - Marcos Pérez-López
- Unit of Toxicology, Veterinary Faculty, University of Extremadura, Caceres, Spain.
| | | | - Antonio González
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain.
| |
Collapse
|
19
|
Huang K, Ma K, Hung Y, Lo L, Lin K, Liu P, Hu M, Chueh S. A new copper ionophore DPMQ protects cells against ultraviolet B irradiation by inhibiting the TRPV1 channel. J Cell Physiol 2018; 233:9594-9610. [DOI: 10.1002/jcp.26861] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/23/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Kuo‐Feng Huang
- Division of Plastic Surgery, Department of Surgery Chi Mei Medical Center Tainan Taiwan Republic of China
| | - Kuo‐Hsing Ma
- Department of Biology and Anatomy National Defense Medical Center Taipei Taiwan Republic of China
| | - Yu‐Chien Hung
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| | - Liang‐Chuan Lo
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| | - Kuo‐Chen Lin
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| | - Pei‐Shan Liu
- Department of Microbiology Soochow University Taipei Taiwan Republic of China
| | - Ming‐Kuan Hu
- Department of Medicinal Chemistry School of Pharmacy, National Defense Medical Center Taipei Taiwan Republic of China
| | - Sheau‐Huei Chueh
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| |
Collapse
|
20
|
Bhatia M. Understanding toxicology: mechanisms and applications. Cell Biol Toxicol 2017; 33:1-4. [PMID: 27714471 DOI: 10.1007/s10565-016-9363-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 09/27/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Madhav Bhatia
- Department of Pathology, University of Otago, Christchurch, 2 Riccarton Avenue, PO Box 4345, Christchurch, 8140, New Zealand.
| |
Collapse
|
21
|
Wang DC, Wang X, Chen C. Effects of anti-human T lymphocyte immune globulins in patients: new or old. J Cell Mol Med 2016; 20:1796-9. [PMID: 27084794 PMCID: PMC4988288 DOI: 10.1111/jcmm.12860] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/29/2016] [Indexed: 02/06/2023] Open
Abstract
Multiple studies demonstrated that anti‐human T lymphocyte immune globulins (ATG) can decrease the incidence of acute and chronic graft rejection in cell or organ transplants. However, further in‐depth study indicates that different subgroups may benefit from either different regimes or alteration of them. Studies among renal transplant patients indicate that low immunological risk patients may not gain the same amount of benefit and thus tilt the risk versus benefit consideration. This may hold true for low immunological risk patients receiving other organ transplants and would be worth further investigation. The recovery time of T cells and natural killer (NK) cells also bears consideration and the impact that it has on the severity and incidence of opportunistic infections closely correlated with the dosage of ATG. The use of lower doses of ATG in combination with other induction medications may offer a solution. The finding that ATG may lose efficacy in cases of multiple transplants or re‐transplants in the case of heart transplants may hold true for other transplantations. This may lead to reconsideration of which induction therapies would be most beneficial in the clinical setting. These studies on ATG done on different patient groups will naturally not be applicable to all, but the evidence accrued from them as a whole may offer us new and different perspectives on how to approach and potentially solve the clinical question of how to best reduce the mortality associated with chronic host‐versus‐graft disease.
Collapse
Affiliation(s)
- Diane C Wang
- Department of Respiratory Medicine, The First Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiangdong Wang
- Department of Respiratory Medicine, The First Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chengshui Chen
- Department of Respiratory Medicine, The First Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
22
|
Guven C, Taskin E, Akcakaya H. Melatonin Prevents Mitochondrial Damage Induced by Doxorubicin in Mouse Fibroblasts Through Ampk-Ppar Gamma-Dependent Mechanisms. Med Sci Monit 2016; 22:438-46. [PMID: 26861593 PMCID: PMC4751923 DOI: 10.12659/msm.897114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Doxorubicin (brand name: Adriamycin®) is used to treat solid tissue cancer but it also affects noncancerous tissues. Its mechanism of cytotoxicity is probably related to increased oxidation, mitochondrial dysfunction, and apoptosis. Melatonin is reported to have antiapoptotic and antioxidative effects. The aim of this study was to determine whether melatonin would counteract in vitro cytotoxicity of doxorubicin in mouse fibroblasts and determine the pathway of its action against doxorubicin-induced apoptosis. MATERIAL AND METHODS We measured markers of apoptosis (cytochrome-c, mitochondrial membrane potential, and apoptotic cell number) and oxidative stress (total oxidant and antioxidant status) and calculated oxidant stress index in 4 groups of fibroblasts: controls, melatonin-treated, doxorubicin-treated, and fibroblasts concomittantly treated with a combination of melatonin and doxorubicin. RESULTS Melatonin given with doxorubicin succesfully countered apoptosis generated by doxorubicin alone, which points to its potential as a protective agent against cell death in doxorubicin chemotherapy. This also implies that patients should be receiving doxorubicin treatment when their physiological level of melatonin is at its highest, which is early in the morning. CONCLUSIONS This physiological level may not be high enough to overcome doxorubicin-induced oxidative stress, but adjuvant melatonin treatment may improve quality of life. Further research is needed to verify our findings.
Collapse
Affiliation(s)
- Celal Guven
- Department of Biophysics, Faculty of Medicine, University of Adiyaman, Adiyaman, Turkey
| | - Eylem Taskin
- Department of Physiotherapy and Rehabilitation, School of Health Sciences, Istanbul Bilim University, Istanbul, Turkey
| | - Handan Akcakaya
- Department of Biophysics, Faculty of Medicine, University of Istanbul, Istanbul, Turkey
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Ever since the first descriptions of methods to isolate pancreatic stellate cells (PSCs) from rodent and human pancreas 17 years ago, rapid advances have been made in our understanding of the biology of these cells and their functions in health and disease. This review updates recent literature in the field, which indicates an increasingly complex role for the cells in normal pancreas, pancreatitis and pancreatic cancer. RECENT FINDINGS Work reported over the past 12 months includes improved methods of PSC immortalization, a role for PSCs in islet fibrosis, novel factors causing PSC activation as well as those inducing quiescence, and translational research aimed at inhibiting the facilitatory effects of PSCs on disease progression in chronic pancreatitis as well as pancreatic cancer. SUMMARY Improved understanding of the role of PSCs in pancreatic pathophysiology has prompted a focus on translational studies aimed at developing novel approaches to modulate PSC function in a bid to improve clinical outcomes of two major fibrotic diseases of the pancreas: chronic pancreatitis and pancreatic cancer.
Collapse
Affiliation(s)
- Minote Apte
- Pancreatic Research Group, South Western Sydney Clinical School, University of New South Wales, and Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia
| | | | | |
Collapse
|