Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(2)2020 Jan 14.
Article in English | MEDLINE | ID: mdl-31947635

ABSTRACT

Hair loss is a disorder in which the hair falls out from skin areas such as the scalp and the body. Several studies suggest the use of herbal medicine to treat related disorders, including alopecia. Dermal microcirculation is essential for hair maintenance, and an insufficient blood supply can lead to hair follicles (HF) diseases. This work aims to provide an insight into the ethnohistorical records of some nutritional compounds containing flavonoids for their potential beneficial features in repairing or recovering from hair follicle disruption. We started from a query for "alopecia" OR "hair loss" AND "Panax ginseng C.A. Mey." (or other six botanicals) terms included in Pubmed and Web of Sciences articles. The activities of seven common botanicals introduced with diet (Panax ginseng C.A. Mey., Malus pumila Mill cultivar Annurca, Coffea arabica, Allium sativum L., Camellia sinensis (L.) Kuntze, Rosmarinum officinalis L., Capsicum annum L.) are discussed, which are believed to reduce the rate of hair loss or stimulate new hair growth. In this review, we pay our attention on the molecular mechanisms underlying the bioactivity of the aforementioned nutritional compounds in vivo, ex vivo and in vitro studies. There is a need for systematic evaluation of the most commonly used plants to confirm their anti-hair loss power, identify possible mechanisms of action, and recommend their best adoption.


Subject(s)
Flavonoids/pharmacology , Hair Follicle/drug effects , Hair Follicle/growth & development , Plant Extracts/pharmacology , Plants, Edible/chemistry , Plants, Medicinal/chemistry , Animals , Flavonoids/chemistry , Flavonoids/metabolism , Humans , Metabolic Networks and Pathways , Molecular Structure , Plant Extracts/chemistry , Plants, Edible/metabolism , Plants, Medicinal/metabolism
2.
J Ethnopharmacol ; 238: 111844, 2019 Jun 28.
Article in English | MEDLINE | ID: mdl-30940568

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Several traditional medicinal herbs are widely used for dermatologic and cosmetic preparations. The beneficial skin repair activity is detected in various phases of wound-healing process, such as cell-cell, cell-matrix interactions or collagen synthesis. AIM OF THE STUDY: The study assessed the effects of Opuntia ficus-indica (L.) Mill. (Opuntia) and Milk Thistle (MT) (Silybum marianum (L.) Gaerth) on adult keratinocytes (HaCaT) functioning under basal condition or in the presence of mechanical damage (wounded cells). MATERIALS AND METHODS: The role of the natural compounds was tested on HaCaT grown in mono-culture and tri-culture configurations. In tri-cultures models, HaCaT were treated with the conditioned media (CM) obtained by Human Normal Dermal Fibroblast (NHDF) and Human Dermal Microvascular Endothelial cells (HMVEC) co-cultures. Specifically, were tested cell viability, oxidative stress mechanisms (cytokines release and lipid peroxidation) and cellular remodelling (growth factors release or metalloproteinase modulation). Moreover, the migratory potential of HaCaT was analysed by the use of wound healing in vitro assay. RESULTS: Opuntia and MT differently modified the metabolism (EGF, MMP-9), and the migratory properties of HaCaT both under physiological conditions or upon mechanical damage (wounded cells). Moreover, both compounds modulated HaCaT response to oxidative stress. The response to the natural compounds were modified, and in some cases potentiated, in tri-culture configuration systems. CONCLUSIONS: The data demonstrated that in vitro tri-culture approach is suitable to characterize the role of natural compounds on the complex communication between dermal-epidermal cellular components and microvascular endothelium. Specifically, Opuntia and MT are good alternatives to synthetic compounds in skin repair promotion.


Subject(s)
Antioxidants/pharmacology , Endothelial Cells/drug effects , Fibroblasts/drug effects , Flavonoids/pharmacology , Keratinocytes/drug effects , Opuntia , Silybum marianum , Cell Culture Techniques , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Coculture Techniques , Culture Media, Conditioned , Diet , Endothelial Cells/physiology , Fibroblasts/physiology , Humans , Keratinocytes/physiology , Wound Healing/drug effects
3.
Nanomedicine (Lond) ; 14(5): 575-594, 2019 03.
Article in English | MEDLINE | ID: mdl-30810075

ABSTRACT

AIM: To clarify the mechanisms of interaction between SiO2 nanoparticles (NPs) and the plasma membrane of GT1-7 neuroendocrine cells, with focus on the activation of calcium-permeable channels, responsible for the long lasting calcium influx and modulation of the electrical activity in these cells. MATERIALS & METHODS: Nontoxic doses of SiO2 NPs were administered to the cells. Calcium imaging and patch clamp techniques were combined with a pharmacological approach. RESULTS: TRPV4, Cx and Panx-like channels are the major components of the NP-induced inward currents. Preincubation with the antioxidant N-acetyl-L-cysteine strongly reduced the [Ca2+]i increase. CONCLUSION: These findings suggest that SiO2 NPs directly activate a complex set of calcium-permeable channels, possibly by catalyzing free radical production.


Subject(s)
Nanoparticles/chemistry , Silicon Dioxide/chemistry , Animals , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Electrophysiology , Lipid Peroxidation/physiology , Mice , Microscopy, Electron, Transmission , Neurons/metabolism , TRPV Cation Channels/metabolism
4.
J Cell Physiol ; 234(5): 7320-7329, 2019 05.
Article in English | MEDLINE | ID: mdl-30417926

ABSTRACT

Current treatments for hair follicle (HF) disruption are based on 5-α reductase inhibitors and prostaglandin modulators. Botanicals and nutraceutical compounds interfere with hair loss or stimulate its partial regrowth. Here, we used in vitro cocultures to investigate the activity of Serenoa repens ( SR) and N-acetyl glucosamine + milk proteins (NAG/Lac) on the paracrine interactions between human microvascular endothelial cells (HMVEC) and HF dermal papilla cells (FDPC). Both SR and NAG/Lac-induced endothelial tubulogenesis were enhanced by FDPC. SR promoted proliferation of both the cell types, while NAG/Lac was effective on endothelium. Vascular endothelial growth factor production, enhanced by SR, was further augmented by FDPC. In FDPC 5-α reductase-II and ß-catenin expressions were modified by SR and less by NAG/Lac, with no additional effect by HMVEC. SR and NAG/Lac prevented lipid peroxidation, whereas NAG/Lac was effective on interleukin 1ß production. Finally, SR and NAG/Lac differentially affected HMVEC permeability and tight junction proteins content. These data provide a mechanistic background for the potential use of these compounds as promoters of HF vascularization.


Subject(s)
Acetylglucosamine/pharmacology , Angiogenesis Inducing Agents/pharmacology , Endothelial Cells/drug effects , Hair Follicle/drug effects , Milk Proteins/pharmacology , Neovascularization, Physiologic/drug effects , Paracrine Communication/drug effects , Plant Extracts/pharmacology , Serenoa , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Endothelial Cells/metabolism , Hair Follicle/cytology , Hair Follicle/metabolism , Humans , Interleukin-1beta/metabolism , Lipid Peroxidation/drug effects , Permeability , Plant Extracts/isolation & purification , Serenoa/chemistry , Signal Transduction , Tight Junctions/drug effects , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
Skin Pharmacol Physiol ; 31(1): 10-18, 2018.
Article in English | MEDLINE | ID: mdl-29131127

ABSTRACT

BACKGROUND: Acne vulgaris is a common skin defect, usually occurring during adolescence, but often it can persist in adults leaving permanent face scarring. Acne is usually treated with topical drugs, oral antibiotics, retinoids, and hormonal therapies, but medicinal plants are increasingly employed. OBJECTIVE: To investigate the protective role of white willow bark (WWB) and 1,2-decanediol (DD) on the damage caused by lipopolysaccharides (LPS) on human adult keratinocytes (HaCaT). METHODS: HaCaT were exposed to LPS alone or in association with WWB and DD. Epidermal viability, metabolic modulation, inflammatory activity, and cell migration were assessed with both common standardized protocols or high-throughput screening systems. RESULTS: The preincubation of HaCaT with WWB and DD (used separately or in combination) differently prevented the alterations induced by LPS on HaCaT in terms of growth factor release (IGF, EGF, VEGF), cytokine production (IL-1α, IL-6, IL-8), or expression of the transcription factor FOXO-I. Moreover, they partially restore wound repair lowered by LPS. CONCLUSIONS: These results suggest that both natural compounds were able to differently affect several functions of LPS-stressed keratinocytes suggesting their potential role for the prevention of acne vulgaris, without adverse effects.


Subject(s)
Glycols/pharmacology , Keratinocytes/drug effects , Plant Bark/chemistry , Salix/chemistry , Acne Vulgaris/drug therapy , Acne Vulgaris/metabolism , Cell Line , Cell Movement/drug effects , Cytokines/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Keratinocytes/metabolism , Lipopolysaccharides/pharmacology , Protective Agents/pharmacology , Skin/drug effects , Wound Healing/drug effects
6.
J Cell Physiol ; 232(2): 243-248, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27334050

ABSTRACT

The molecular and cellular mechanisms underlying vascular remodeling are currently investigated by experimental strategies which aim to mimic the complex environmental conditions found in vivo. Some of them focus on the tubulogenic activity of dispersed endothelial cell populations, while others evaluate vascular sprouting. Here we propose a new method to assess matrigel invasion starting from confluent or subconfluent monolayers of human microvascular ECs (HMVEC) seeded on different substrates. The experimental setting is also validated by an improved hybrid multiscale mathematical approach, which integrates a mesoscopic grid-based cellular Potts model, that describes HMVEC phenomenology, with a continuous one, accounting for the kinetics of diffusing growth factors. Both experimental and theoretical approaches show that the endothelial potential to invade, migrate, and organize in tubule structures is a function of selected environmental parameters. The present methodology is intended to be simple to use, standardized for rapid screening and suitable for mechanistic studies. J. Cell. Physiol. 232: 243-248, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Biological Assay/methods , Computer Simulation , Endothelial Cells/cytology , Inventions , Cell Line , Humans , Microvessels/cytology
7.
J Cell Physiol ; 232(5): 897-903, 2017 05.
Article in English | MEDLINE | ID: mdl-27764901

ABSTRACT

The biological importance of circulatory blood supply and angiogenesis for hair growth is now well recognized, but the their regulatory mechanisms require more mechanistic investigation. In vitro cocultures and tricultures can be successfully employed to greatly improve our knowledge on paracrine crosstalk between cell types that populate the dermal-epidermal interface and cutaneous vasculature. Here we report that human dermal fibroblasts (NHDF) promote viability and proliferation of microvascular endothelial cells (HMVEC), while HMVEC are not mitogenic for NHDF. In triculture setup, conditioned media (CM) obtained by cocultures (HMVEC/NHDF or HMVEC/follicle fibroblasts) differently modulate growth and proliferation of keratinocytes and alter the expression of metabolic and pro-inflammatory markers. In conclusion, tricultures were successfully employed to characterize in vitro dermal-epithelial and endothelial interactions and could integrate ex vivo and in vivo approaches by the use of high-throughput and standardized protocols in controlled conditions. J. Cell. Physiol. 232: 897-903, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Communication , Dermis/cytology , Endothelial Cells/cytology , Epidermal Cells , Microvessels/cytology , Adult , Biomarkers/metabolism , Cell Communication/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media, Conditioned/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Inflammation/pathology , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Wound Healing/drug effects
8.
Nat Prod Res ; 30(24): 2831-2834, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26936689

ABSTRACT

Some natural compounds, including flavonoids, are active in vasculature re-growth during hair follicle disruption, but their effects have not been yet evaluated directly on microvascular endothelial cells. Skin vascularisation regulates the physiological blood supply required for hair growth and its dysregulation is the basis of several human diseases. Follicle-derived vascular endothelial growth factor (VEGF) release from follicular keratinocytes promotes perifollicular vascularisation and increases follicle and hair size, while blockade of VEGF-mediated angiogenesis leads to impaired hair growth. Here, we tested three flavonoids, namely visnadin (VSD), hesperidin (HSP) and baicalin (BC), on cultured human microvascular endothelial cells (HMEC), comparing their effects with minoxidil (MXD), a synthetic drug broadly used in the treatment of androgenetic alopecia. The response to these compounds was assayed in terms of endothelial survival, proliferation, tubulogenesis and proangiogenic signalling. We show that BC promotes HMEC proliferation, while both VSD and MXD enhance tubulogenesis. Interestingly, only HSP increases VEGFR-2 phosphorylation.

10.
Comput Biol Med ; 63: 143-56, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26079199

ABSTRACT

Angiogenesis, the formation of new blood vessel networks from existing capillary or post-capillary venules, is an intrinsically multiscale process occurring in several physio-pathological conditions. In particular, hypoxic tissue cells activate downstream cascades culminating in the secretion of a wide range of angiogenic factors, including VEGF isoforms. Such diffusive chemicals activate the endothelial cells (ECs) forming the external walls of the nearby vessels that chemotactically migrate toward the hypoxic areas of the tissue as multicellular sprouts. A functional network eventually emerges by further branching and anastomosis processes. We here propose a CPM-based approach reproducing selected features of the angiogenic progression necessary for the reoxygenation of a hypoxic tissue. Our model is able to span the different scale involved in the angiogenic progression as it incorporates reaction-diffusion equations for the description of the evolution of microenvironmental variables in a discrete mesoscopic cellular Potts model (CPM) that reproduces the dynamics of the vascular cells. A key feature of this work is the explicit phenotypic differentiation of the ECs themselves, distinguished in quiescent, stalk and tip. The simulation results allow identifying a set of key mechanisms underlying tissue vascularization. Further, we provide evidence that the nascent pattern is characterized by precise topological properties. Finally, we link abnormal sprouting angiogenesis with alteration in selected cell behavior.


Subject(s)
Cell Differentiation , Endothelial Cells/metabolism , Hypoxia , Models, Cardiovascular , Neovascularization, Physiologic , Animals , Humans , Hypoxia/metabolism , Hypoxia/physiopathology , Protein Isoforms/metabolism , Vascular Endothelial Growth Factor A/metabolism
11.
PLoS One ; 10(3): e0119790, 2015.
Article in English | MEDLINE | ID: mdl-25774921

ABSTRACT

Catestatin (Cst) is a 21-amino acid peptide deriving from Chromogranin A. Cst exerts an overall protective effect against an excessive sympathetic stimulation of cardiovascular system, being able to antagonize catecholamine secretion and to reduce their positive inotropic effect, by stimulating the release of nitric oxide (NO) from endothelial cells. Moreover, Cst reduces ischemia/reperfusion (I/R) injury, improving post-ischemic cardiac function and cardiomyocyte survival. To define the cardioprotective signaling pathways activated by Cst (5 nM) we used isolated adult rat cardiomyocytes undergoing simulated I/R. We evaluated cell viability rate with propidium iodide labeling and mitochondrial membrane potential (MMP) with the fluorescent probe JC-1. The involvement of Akt, GSK3ß, eNOS and phospholamban (PLN) cascade was studied by immunofluorescence. The role of PI3K-Akt/NO/cGMP pathway was also investigated by using the pharmacological blockers wortmannin (Wm), L-NMMA and ODQ. Our experiments revealed that Cst increased cell viability rate by 65% and reduced cell contracture in I/R cardiomyocytes. Wm, L-NMMA and ODQ limited the protective effect of Cst. The protective outcome of Cst was related to its ability to maintain MMP and to increase AktSer473, GSK3ßSer9, PLNThr17 and eNOSSer1179 phosphorylation, while treatment with Wm abolished these effects. Thus, the present results show that Cst is able to exert a direct action on cardiomyocytes and give new insights into the molecular mechanisms involved in its protective effect, highlighting the PI3K/NO/cGMP pathway as the trigger and the MMP preservation as the end point of its action.


Subject(s)
Chromogranin A/pharmacology , Membrane Potential, Mitochondrial/drug effects , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Peptide Fragments/pharmacology , Signal Transduction/drug effects , Animals , Cell Survival/drug effects , Chromogranin A/therapeutic use , Glycogen Synthase Kinase 3/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Peptide Fragments/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats
12.
Exp Dermatol ; 24(5): 388-90, 2015 May.
Article in English | MEDLINE | ID: mdl-25690790

ABSTRACT

Human follicle dermal papilla cells (FDPC) are a specialized population of mesenchymal cells located in the skin. They regulate hair follicle (HF) development and growth, and represent a reservoir of multipotent stem cells. Growing evidence supports the hypothesis that HF cycling is associated with vascular remodeling. Follicular keratinocytes release vascular endothelial growth factor (VEGF) that sustains perifollicular angiogenesis leading to an increase of follicle and hair size. Furthermore, several human diseases characterized by hair loss, including Androgenetic Alopecia, exhibit alterations of skin vasculature. However, the molecular mechanisms underlying HF vascularization remain largely unknown. In vitro coculture approaches can be successfully employed to greatly improve our knowledge and shed more light on this issue. Here we used Transwell-based co-cultures to show that FDPC promote survival, proliferation and tubulogenesis of human microvascular endothelial cells (HMVEC) more efficiently than fibroblasts. Accordingly, FDPC enhance the endothelial release of VEGF and IGF-1, two well-known proangiogenic growth factors. Collectively, our data suggest a key role of papilla cells in vascular remodeling of the hair follicle.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Hair Follicle/cytology , Hair Follicle/metabolism , Cell Proliferation , Cell Survival , Coculture Techniques , Hair/growth & development , Hair Follicle/blood supply , Humans , Insulin-Like Growth Factor I/metabolism , Interleukin-1alpha/biosynthesis , Neovascularization, Physiologic , Paracrine Communication , Vascular Endothelial Growth Factor A/metabolism , Vascular Remodeling , beta Catenin/biosynthesis
13.
Biomed Res Int ; 2014: 783623, 2014.
Article in English | MEDLINE | ID: mdl-25136621

ABSTRACT

The chromogranin-A peptide catestatin modulates a wide range of processes, such as cardiovascular functions, innate immunity, inflammation, and metabolism. We recently found that the cardiac antiadrenergic action of catestatin requires a PI3K-dependent NO release from endothelial cells, although the receptor involved is yet to be identified. In the present work, based on the cationic properties of catestatin, we tested the hypothesis of its interaction with membrane heparan sulphate proteoglycans, resulting in the activation of a caveolae-dependent endocytosis. Experiments were performed on bovine aortic endothelial cells. Endocytotic vesicles trafficking was quantified by confocal microscopy using a water-soluble membrane dye; catestatin colocalization with heparan sulphate proteoglycans and caveolin 1 internalization were studied by fluorimetric measurements in live cells. Modulation of the catestatin-dependent eNOS activation was assessed by immunofluorescence and immunoblot analysis. Our results demonstrate that catestatin (5 nM) colocalizes with heparan sulphate proteoglycans and induces a remarkable increase in the caveolae-dependent endocytosis and caveolin 1 internalization, which were significantly reduced by both heparinase and wortmannin. Moreover, catestatin was unable to induce Ser(1179) eNOS phosphorylation after pretreatments with heparinase and methyl-ß-cyclodextrin. Taken together, these results highlight the obligatory role for proteoglycans and caveolae internalization in the catestatin-dependent eNOS activation in endothelial cells.


Subject(s)
Chromogranin A/administration & dosage , Endocytosis/drug effects , Heparan Sulfate Proteoglycans/metabolism , Nitric Oxide/metabolism , Peptide Fragments/administration & dosage , Animals , Aorta/drug effects , Aorta/metabolism , Cattle , Caveolae/drug effects , Caveolae/ultrastructure , Caveolin 1/metabolism , Chromogranin A/metabolism , Chromogranin A/ultrastructure , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Nitric Oxide Synthase Type III/metabolism , Peptide Fragments/metabolism , Peptide Fragments/ultrastructure , Transport Vesicles/metabolism , Transport Vesicles/ultrastructure
14.
Cardiovasc Diabetol ; 11: 129, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-23066908

ABSTRACT

BACKGROUND: The aim of this study was to investigate whether obestatin (OB), a peptide mediator encoded by the ghrelin gene exerting a protective effect in ischemic reperfused heart, is able to reduce cardiac dysfunctions in adult diabetic rats. METHODS: Diabetes was induced by STZ injection (50 mg/kg) in Wistar rats (DM). OB was administered (25 µg/kg) twice a day for 6 weeks. Non-diabetic (ND) rats and DM rats were distributed into four groups: untreated ND, OB-treated ND, untreated DM, OB-treated DM. Cardiac contractility and ß-adrenergic response were studied on isolated papillary muscles. Phosphorylation of AMPK, Akt, ERK1/2 and GSK3ß as well ß-1 adrenoreceptors levels were detected by western blot, while α-MHC was measured by RT-PCR. RESULTS: OB preserved papillary muscle contractility (85 vs 27% of ND), ß-adrenergic response (103 vs 65% of ND), as well ß1-adrenoreceptors and α-MHC levels in diabetic myocardial tissue. Moreover, OB up-regulated the survival kinases Akt and ERK1/2, and enhanced AMPK and GSK3ß phosphorylation. OB corrected oxidative unbalance, reduced pro-inflammatory cytokine TNF-α plasma levels, NFkB translocation and pro-fibrogenic factors expression in diabetic myocardium. CONCLUSIONS: OB displays a significant beneficial effect against the alterations of contractility and ß-adrenergic response in the heart of STZ-treated diabetic rats, which was mainly associated with the ability of OB to up-regulate the transcription of ß1-adrenergic receptors and α-MHC; this protective effect was accompanied by the ability to restore oxidative balance and to promote phosphorylation/modulation of AMPK and pro-survival kinases such as Akt, ERK1/2 and GSK3ß.


Subject(s)
Cardiotonic Agents/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Heart Diseases/drug therapy , Myocardial Contraction/drug effects , Papillary Muscles/drug effects , Peptide Hormones/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Cell Line , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Heart Diseases/etiology , Heart Diseases/metabolism , Heart Diseases/physiopathology , Hypoglycemic Agents/pharmacology , Inflammation Mediators/blood , Male , Metformin/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Papillary Muscles/metabolism , Papillary Muscles/physiopathology , Phosphorylation , Polymerase Chain Reaction , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, beta-1/metabolism , Recovery of Function , Time Factors , Tumor Necrosis Factor-alpha/blood , Ventricular Myosins/genetics , Ventricular Myosins/metabolism
15.
FASEB J ; 26(7): 2888-98, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22459152

ABSTRACT

Three forms of serpinin peptides, serpinin (Ala26Leu), pyroglutaminated (pGlu)-serpinin (pGlu23Leu), and serpinin-Arg-Arg-Gly (Ala29Gly), are derived from cleavage at pairs of basic residues in the highly conserved C terminus of chromogranin A (CgA). Serpinin induces PN-1 expression in neuroendocrine cells to up-regulate granule biogenesis via a cAMP-protein kinase A-Sp1 pathway, while pGlu-serpinin inhibits cell death. The aim of this study was to test the hypothesis that serpinin peptides are produced in the heart and act as novel ß-adrenergic-like cardiac modulators. We detected serpinin peptides in the rat heart by HPLC and ELISA methods. The peptides included predominantly Ala29Gly and pGlu-serpinin and a small amount of serpinin. Using the Langendorff perfused rat heart to evaluate the hemodynamic changes, we found that serpinin and pGlu-serpinin exert dose-dependent positive inotropic and lusitropic effects at 11-165 nM, within the first 5 min after administration. The pGlu-serpinin-induced contractility is more potent than that of serpinin, starting from 1 nM. Using the isolated rat papillary muscle preparation to measure contractility in terms of tension development and muscle length, we further corroborated the pGlu-serpinin-induced positive inotropism. Ala29Gly was unable to affect myocardial performance. Both pGlu-serpinin and serpinin act through a ß1-adrenergic receptor/adenylate cyclase/cAMP/PKA pathway, indicating that, contrary to the ß-blocking profile of the other CgA-derived cardiosuppressive peptides, vasostatin-1 and catestatin, these two C-terminal peptides act as ß-adrenergic-like agonists. In cardiac tissue extracts, pGlu-serpinin increased intracellular cAMP levels and phosphorylation of phospholamban (PLN)Ser16, ERK1/2, and GSK-3ß. Serpinin and pGlu-serpinin peptides emerge as novel ß-adrenergic inotropic and lusitropic modulators, suggesting that CgA and the other derived cardioactive peptides can play a key role in how the myocardium orchestrates its complex response to sympathochromaffin stimulation.


Subject(s)
Adrenergic beta-1 Receptor Agonists/chemistry , Adrenergic beta-1 Receptor Agonists/pharmacology , Cardiotonic Agents/chemistry , Cardiotonic Agents/pharmacology , Chromogranin A/chemistry , Chromogranin A/physiology , Heart/drug effects , Heart/physiology , Peptide Fragments/chemistry , Peptide Fragments/physiology , Amino Acid Sequence , Amino Acid Substitution , Animals , Chromogranin A/genetics , Chromogranin A/pharmacology , In Vitro Techniques , Male , Molecular Sequence Data , Myocardial Contraction/drug effects , Myocardium/chemistry , Papillary Muscles/drug effects , Papillary Muscles/physiology , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Rats , Rats, Wistar
16.
Cardiovasc Res ; 91(4): 617-24, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21543385

ABSTRACT

AIMS: Catestatin (CST) is a chromogranin A (CgA)-derived peptide (hCgA352-372) with three identified human variants (G364S/P370L/R374Q-CST) that show differential potencies towards the inhibition of catecholamine release. Although CST affects several cardiovascular parameters, the mechanisms underlying CST action in the heart have remained elusive. Therefore, we sought to determine the mechanism of action of CST and its variants on ventricular myocardium and endothelial cells. METHODS AND RESULTS: Contractile force and Ca(2+) transients were measured, respectively, on rat papillary muscles and isolated cardiomyocytes (CC) under basal conditions and after ß-adrenergic stimulation. Nitric oxide (NO) production and endothelial nitric oxide synthase (eNOS) phosphorylation (P(Ser1179)eNOS) were studied in bovine aortic endothelial (BAE-1) cells. Under basal conditions, wild-type CST (WT-CST, 10-50 nM) transiently enhanced myocardial contractility. CST variants (G364S and P370L) exerted a comparable positive inotropic effect. The H(1) histamine receptor antagonist mepyramine abolished the increase of contractile force induced by WT-CST. Moreover, WT-CST dose-dependently (5-50 nM) reduced the effect of ß-adrenergic stimulation. This anti-adrenergic effect was not mediated by a direct action on CC, but involved a PI3K-dependent NO release from endocardial endothelial cells. Indeed, CST induced a wortmannin-sensitive, Ca(2+)-independent increase in NO production and eNOS phosphorylation on BAE-1 cells. While the anti-adrenergic and NO release effects of P370L-CST were comparable with those of WT-CST, the G364S variant was ineffective on the same parameters. CONCLUSION: Our results suggest that the anti-adrenergic action of CST depends on the endothelial PI3K-Akt-eNOS pathway and that its structural alterations entail functional features that correlate with the different anti-hypertensive potential described in humans.


Subject(s)
Adrenergic Antagonists/pharmacology , Chromogranin A/pharmacology , Heart/drug effects , Nitric Oxide Synthase Type III/physiology , Peptide Fragments/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/drug effects , Animals , Blotting, Western , Calcium/metabolism , Cattle , Cells, Cultured , Fluorescent Antibody Technique , Myocardium/metabolism , Nitric Oxide/biosynthesis , Papillary Muscles/drug effects , Rats
17.
Exp Biol Med (Maywood) ; 236(4): 390-401, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21378031

ABSTRACT

The present review is focused on the dual role played by platelet-activating factor (PAF) in ischemia and reperfusion (I/R) injury of the heart. Although the involvement of PAF in the pathogenesis of myocardial reperfusion injury is well established, in the last few years it has emerged that very low concentrations of PAF exert cardioprotective effects, comparable to that afforded by ischemic preconditioning (IP). PAF is a potent phosphoglyceride involved in different pathophysiological conditions affecting the cardiovascular system, including the development of myocardial I/R injury. PAF is released from the I/R myocardium in concentrations (1-10 nmol/L) high enough to negatively modulate coronary circulation as well as electrical and contractile activities. PAF may act either directly, via generation of secondary mediators, or through the activation of inflammatory cells like platelets and polymorphonuclear neutrophils, which exacerbate postischemic myocardial injury. The effects of PAF are mediated through specific receptors (PAFRs) that belong to the superfamily of G protein-coupled receptors. Since cardiomyocytes not only produce PAF but also possess PAFRs, it is likely that PAF acts as an autocrine/paracrine mediator. Although the negative effects exerted by high concentrations of PAF are well established, several recent findings from our and other laboratories have demonstrated that very low concentrations (pmol/L) of PAF infused before ischemia induce cardioprotective effects similar to those afforded by IP, and that endogenous PAF production participates in the induction of IP itself. The IP-like action exerted by low concentrations of PAF is due to the activation/phosphorylation of kinases included in the reperfusion injury salvage kinase (RISK) pathway, such as protein kinase C, Akt/PkB and nitric oxide synthase. Together with the activation of mitochondrial K(ATP) channels, these events may allow prevention of mitochondrial permeability transition pores opening at reperfusion. Moreover, the nitric oxide-dependent S-nitrosylation of L-type Ca(2+) channels induced by PAF reduces intracellular Ca(2+) overload.


Subject(s)
Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/physiopathology , Platelet Activating Factor/physiology , Humans , Ischemic Preconditioning , Platelet Activating Factor/biosynthesis
18.
Cell Mol Neurobiol ; 30(8): 1171-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21104119

ABSTRACT

The Chromogranin A (CgA)-derived anti-hypertensive peptide catestatin (CST) antagonizes catecholamine secretion, and is a negative myocardial inotrope acting via a nitric oxide-dependent mechanism. It is not known whether CST contributes to ischemia/reperfusion injury or is a component of a cardioprotective response to limit injury. Here, we tested whether CST by virtue of its negative inotropic activity improves post-ischemic cardiac function and cardiomyocyte survival. Three groups of isolated perfused hearts from adult Wistar rats underwent 30-min ischemia and 120-min reperfusion (I/R, Group 1), or were post-conditioned by brief ischemic episodes (PostC, 5-cycles of 10-s I/R at the beginning of 120-min reperfusion, Group 2), or with exogenous CST (75 nM for 20 min, CST-Post, Group-3) at the onset of reperfusion. Perfusion pressure and left ventricular pressure (LVP) were monitored. Infarct size was evaluated with nitroblue-tetrazolium staining. The CST (5 nM) effects were also tested in simulated ischemia/reperfusion experiments on cardiomyocytes isolated from young-adult rats, evaluating cell survival with propidium iodide labeling. Infarct size was 61 ± 6% of risk area in hearts subjected to I/R only. PostC reduced infarct size to 34 ± 5%. Infarct size in CST-Post was 36 ± 3% of risk area (P < 0.05 respect to I/R). CST-Post reduced post-ischemic rise of diastolic LVP, an index of contracture, and significantly improved post-ischemic recovery of developed LVP. In isolated cardiomyocytes, CST increased the cell viability rate by about 65% after simulated ischemia/reperfusion. These results suggest a novel cardioprotective role for CST, which appears mainly due to a direct reduction of post-ischemic myocardial damages and dysfunction, rather than to an involvement of adrenergic terminals and/or endothelium.


Subject(s)
Chromogranin A/pharmacology , Heart/physiopathology , Myocardial Ischemia/drug therapy , Myocardial Ischemia/physiopathology , Peptide Fragments/pharmacology , Reperfusion Injury/drug therapy , Reperfusion Injury/physiopathology , Ventricular Function, Left/drug effects , Animals , Cell Separation , Cell Survival , Chromogranin A/therapeutic use , Diastole/drug effects , Heart Function Tests/drug effects , In Vitro Techniques , Male , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Ischemia/complications , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Peptide Fragments/therapeutic use , Rats , Rats, Wistar , Reperfusion Injury/complications , Systole/drug effects
19.
Am J Physiol Heart Circ Physiol ; 299(2): H470-81, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20525876

ABSTRACT

Obestatin, a newly discovered peptide encoded by the ghrelin gene, induces the expression of genes regulating pancreatic beta-cell differentiation, insulin biosynthesis, and glucose metabolism. It also activates antiapoptotic signaling pathways such as phosphoinositide 3-kinase (PI3K) and ERK1/2 in pancreatic beta-cells and human islets. Since these kinases have been shown to protect against myocardial injury, we sought to investigate whether obestatin would exert cardioprotective effects. Both isolated perfused rat heart and cultured cardiomyocyte models of ischemia-reperfusion (I/R) were used to measure infarct size and cell apoptosis as end points of injury. The presence of specific obestatin receptors on cardiac cells as well as the signaling pathways underlying the obestatin effect were also studied. In the isolated heart, the addition of rat obestatin-(1-23) before ischemia reduced infarct size and contractile dysfunction in a concentration-dependent manner, whereas obestatin-(23-1), a synthetic analog with an inverse aminoacid sequence, was ineffective. The cardioprotective effect of obestatin-(1-23) was observed at concentrations of 10-50 nmol/l and was abolished by inhibiting PI3K or PKC by the addition of wortmannin (100 nmol/l) or chelerythrine, (5 micromol/l), respectively. In rat H9c2 cardiac cells or isolated ventricular myocytes subjected to I/R, 50 nmol/l obestatin-(1-23) reduced cardiomyocyte apoptosis and reduced caspase-3 activation; the antiapoptotic effect was blocked by the inhibition of PKC, PI3K, or ERK1/2 pathways. In keeping with these functional findings, radioreceptor binding results revealed the presence of specific high-affinity obestatin-binding sites, mainly localized on membranes of the ventricular myocardium and cardiomyocytes. Our data suggest that, by acting on specific receptors, obestatin-(1-23) activates PI3K, PKC-epsilon, PKC-delta, and ERK1/2 signaling and protects cardiac cells against myocardial injury and apoptosis induced by I/R.


Subject(s)
Apoptosis , Myocardial Contraction , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Peptide Hormones/metabolism , Ventricular Function, Left , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Peptide Hormones/administration & dosage , Peptides/pharmacology , Perfusion , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Receptors, Ghrelin/metabolism , Signal Transduction , Time Factors , Ventricular Function, Left/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...