1
|
Yang L, Wang G, Ma Y, Zhao Q, Zhao H, Wang Q, Zhong C, Zhang C, Yang Y. TRPML1 acts as a predisposing factor in lymphedema development by regulating the subcellular localization of aquaporin-3, -5. PLoS One 2024; 19:e0310653. [PMID: 39637010 PMCID: PMC11620549 DOI: 10.1371/journal.pone.0310653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/04/2024] [Indexed: 12/07/2024] Open
Abstract
An imbalance in lymphatic fluid, whether it is caused by generation, transport, outflow, or dysfunctional vessels, can lead to lymphedema; however, the exact pathogenesis of this disease remains unclear. To explore the mechanism, we focused on the association among TRPML1, aquaporin-3 (AQP3), and aquaporin-5 (AQP5) in human lymphatic endothelial cells (HLECs). We explored the role of TRPML1 in altering the permeability of HLECs in lymphedema. Meanwhile, we constructed a disease model using gene-knockout mice to observe the effect of TRPML1 on inflammation and fibrosis in lymphedema sites. Our results indicate that TRPML1 not only regulates the localization of AQP3, -5 to the cell membrane but also increases HLEC permeability, disrupts lymphatic fluid transport, and mediates the development of chronic inflammation at the site of lymphedema. Our study suggests that TRPML1 is a precipitating factor in lymphedema. Our findings improve the understanding of TRPML1 and aquaporins in secondary lymphedema, providing valuable insights for future research.
Collapse
Affiliation(s)
- Lijie Yang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Guanzheng Wang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Yuan Ma
- College of Basic Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Qiancheng Zhao
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - He Zhao
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Qi Wang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Chonghua Zhong
- College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Chunmei Zhang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Yiming Yang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
- College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
2
|
Filippelli A, Ciccone V, Del Gaudio C, Simonis V, Frosini M, Tusa I, Menconi A, Rovida E, Donnini S. ERK5 mediates pro-tumorigenic phenotype in non-small lung cancer cells induced by PGE2. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119810. [PMID: 39128596 DOI: 10.1016/j.bbamcr.2024.119810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/25/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, with non-small cell lung cancer (NSCLC) constituting approximately 84 % of all lung cancer cases. The role of inflammation in the initiation and progression of NSCLC tumors has been the focus of extensive research. Among the various inflammatory mediators, prostaglandin E2 (PGE2) plays a pivotal role in promoting the aggressiveness of epithelial tumors through multiple mechanisms, including the stimulation of growth, evasion of apoptosis, invasion, and induction of angiogenesis. The Extracellular signal-Regulated Kinase 5 (ERK5), the last discovered member among conventional mitogen-activated protein kinases (MAPK), is implicated in cancer-associated inflammation. In this study, we explored whether ERK5 is involved in the process of tumorigenesis induced by PGE2. Using A549 and PC9 NSCLC cell lines, we found that PGE2 triggers the activation of ERK5 via the EP1 receptor. Moreover, both genetic and pharmacological inhibition of ERK5 reduced PGE2-induced proliferation, migration, invasion and stemness of A549 and PC9 cells, indicating that ERK5 plays a critical role in PGE2-induced tumorigenesis. In summary, our study underscores the pivotal role of the PGE2/EP1/ERK5 axis in driving the malignancy of NSCLC cells in vitro. Targeting this axis holds promise as a potential avenue for developing novel therapeutic strategies aimed at controlling the advancement of NSCLC.
Collapse
Affiliation(s)
| | - Valerio Ciccone
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Cinzia Del Gaudio
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Vittoria Simonis
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Maria Frosini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessio Menconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy.
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| |
Collapse
|
3
|
Ciccone V, Simonis V, Del Gaudio C, Cucini C, Ziche M, Morbidelli L, Donnini S. ALDH1A1 confers resistance to RAF/MEK inhibitors in melanoma cells by maintaining stemness phenotype and activating PI3K/AKT signaling. Biochem Pharmacol 2024; 224:116252. [PMID: 38701866 DOI: 10.1016/j.bcp.2024.116252] [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: 11/24/2023] [Revised: 04/16/2024] [Accepted: 04/30/2024] [Indexed: 05/05/2024]
Abstract
The mitogen-activated protein kinase (MAPK/ERK) pathway is pivotal in controlling the proliferation and survival of melanoma cells. Several mutations, including those in BRAF, exhibit an oncogenic effect leading to increased cellular proliferation. As a result, the combination therapy of a MEK inhibitor with a BRAF inhibitor demonstrated higher efficacy and lower toxicity than BRAF inhibitor alone. This combination has become the preferred standard of care for tumors driven by BRAF mutations. Aldehyde dehydrogenase 1A1 (ALDH1A1) is a known marker of stemness involved in drug resistance in several type of tumors, including melanoma. This study demonstrates that melanoma cells overexpressing ALDH1A1 displayed resistance to vemurafenib and trametinib through the activation of PI3K/AKT signaling instead of MAPK axis. Inhibition of PI3K/AKT signaling partially rescued sensitivity to the drugs. Consistently, pharmacological inhibition of ALDH1A1 activity downregulated the activation of AKT and partially recovered responsiveness to vemurafenib and trametinib. We propose ALDH1A1 as a new potential target for treating melanoma resistant to MAPK/ERK inhibitors.
Collapse
Affiliation(s)
- Valerio Ciccone
- Department of Life Sciences, University of Siena, Siena I-53100, Italy
| | - Vittoria Simonis
- Department of Life Sciences, University of Siena, Siena I-53100, Italy
| | - Cinzia Del Gaudio
- Department of Life Sciences, University of Siena, Siena I-53100, Italy
| | - Claudio Cucini
- Department of Life Sciences, University of Siena, Siena I-53100, Italy
| | - Marina Ziche
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena I‑53100, Italy
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Siena I-53100, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Siena I-53100, Italy.
| |
Collapse
|
4
|
Genah S, Ciccone V, Filippelli A, Simonis V, Martelli A, Piragine E, Pagnotta E, Pecchioni N, Calderone V, Morbidelli L. Erucin, a natural isothiocyanate, exerts pro-angiogenic properties in cultured endothelial cells and reverts angiogenic impairment induced by high glucose. Phytother Res 2024; 38:2641-2655. [PMID: 38488442 DOI: 10.1002/ptr.8183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/27/2023] [Accepted: 02/21/2024] [Indexed: 06/13/2024]
Abstract
Insufficient vessel maintenance adversely impacts patients in terms of tissue reperfusion following stroke or myocardial infarction, as well as during wound healing. Angiogenesis impairment is a feature typical of metabolic disorders acting at the cardiovascular level, such as diabetes. Therapeutic angiogenesis regulation offers promising clinical implications, and natural compounds as pro-angiogenic nutraceuticals hold valuable applications in regenerative medicine. By using cultured endothelial cells from human umbilical veins (HUVEC) we studied functional and molecular responses following exposure to erucin, a natural isothiocyanate derived from Brassicaceae plants and extracted from the seeds of rocket. Erucin (at nanomolar concentrations) promotes cell migration and tube formation, similar to vascular endothelial growth factor (VEGF), through mobilizing paxillin at endothelial edges. At the molecular level, erucin induces signaling pathways typical of angiogenesis activation, namely Ras, PI3K/AKT, and ERK1/2, leading to VEGF expression and triggering its autocrine production, as pharmacological inhibition of soluble VEGF and VEGFR2 dampens endothelial functions. Furthermore, erucin, alone and together with VEGF, preserves endothelial angiogenic functions under pathological conditions, such as those induced in HUVEC by high glucose (HG) exposure. Erucin emerges as a compelling candidate for therapeutic revascularization applications, showcasing promising prospects for natural compounds in regenerative medicine, particularly in addressing angiogenesis-related disorders.
Collapse
Affiliation(s)
- Shirley Genah
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Valerio Ciccone
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | | | - Alma Martelli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Eleonora Pagnotta
- Research Centre for Cereal and Industrial Crops, CREA, Bologna, Italy
| | - Nicola Pecchioni
- Research Centre for Cereal and Industrial Crops, CREA, Foggia, Italy
| | | | | |
Collapse
|
5
|
Ciccone V, Terzuoli E, Ristori E, Filippelli A, Ziche M, Morbidelli L, Donnini S. ALDH1A1 overexpression in melanoma cells promotes tumor angiogenesis by activating the IL‑8/Notch signaling cascade. Int J Mol Med 2022; 50:99. [PMID: 35656893 PMCID: PMC9186295 DOI: 10.3892/ijmm.2022.5155] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/29/2022] [Indexed: 11/06/2022] Open
Abstract
ALDH1A1 is a cytosolic enzyme upregulated in tumor cells, involved in detoxifying cells from reactive aldehydes and in acquiring resistance to chemotherapeutic drugs. Its expression correlates with poor clinical outcomes in a number of cancers, including melanoma. The present study hypothesized that the increased ALDH1A1 expression and activity upregulated the release of proangiogenic factors from melanoma cells, which regulate angiogenic features in endothelial cells (ECs) through a rearrangement of the Notch pathway. In vivo, when subcutaneously implanted in immunodeficient mice, ALDH1A1 overexpressing melanoma cells displayed a higher microvessel density. In a 3D multicellular system, obtained co‑culturing melanoma cancer cells with stromal cells, including ECs, melanoma ALDH1A1 overexpression induced the recruitment of ECs into the core of the tumorspheres. By using a genes array, overexpression of ALDH1A1 in tumor cells also promoted modulation of Notch cascade gene expression in ECs, suggesting an interaction between tumor cells and ECs mediated by enrichment of angiogenic factors in the tumor microenvironment. To confirm this hypothesis, inactivation of ALDH1A1 by the pharmacological inhibitor CM037 significantly affected the release of angiogenic factors, including IL‑8, from melanoma cells. High levels of ALDH1A1, through the retinoic acid pathway, regulated the activation of NF‑kB‑p65 and IL‑8. Further, in a 2D co‑culture system, the addition of an IL‑8 neutralizing antibody to ECs co‑cultured with melanoma cells forced to express ALDH1A1 dampened endothelial angiogenic features, both at the molecular (in terms of gene and protein expression of mediators of the Notch pathway) and at the functional level (proliferation, scratch assay, tube formation and permeability). In conclusion, these findings demonstrated the existence of a link between melanoma ALDH1A1 expression and EC Notch signaling modification that results in a pro‑angiogenic phenotype. Based on the crucial role of ALDH1A1 in melanoma control of the tumor microenvironment, the enzyme seems a promising target for the development of novel drugs able to interrupt the cross‑talk between cancer (stem) cells and endothelial cells.
Collapse
Affiliation(s)
- Valerio Ciccone
- Department of Life Sciences, University of Siena, Siena I‑53100, Italy
| | - Erika Terzuoli
- Department of Life Sciences, University of Siena, Siena I‑53100, Italy
| | - Emma Ristori
- Department of Life Sciences, University of Siena, Siena I‑53100, Italy
| | | | - Marina Ziche
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena I‑53100, Italy
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Siena I‑53100, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Siena I‑53100, Italy
| |
Collapse
|
6
|
Characterization of the Safety Profile of Sweet Chestnut Wood Distillate Employed in Agriculture. SAFETY 2021. [DOI: 10.3390/safety7040079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
In organic agriculture, synthetic pesticides and treatments are substituted by natural remedies with interesting success for product yield and environmental outcomes, but the safety of these bio-based products needs to be assessed in vertebrate and human models. Therefore, in this paper we assessed the safety profile of sweet chestnut (Castanea sativa) wood distillate (WD) on the different cellular components of tissues implied in transcutaneous absorption. We investigated the viability of different cell lines mimicking the skin (HaCaT keratinocytes), mucosa (A431), connective (normal human dermal fibroblasts, NHDF) and vascular (human umbilical vein endothelial cells, HUVEC) tissues after exposure to increasing concentrations (0.04–0.5%, v/v, corresponding to 1:2800–1:200 dilutions) of WD. A short exposure to increasing doses of WD was well tolerated up to the highest concentration. Instead, following a prolonged treatment, a concentration dependent cytotoxic effect was observed. Notably, a different behavior was found with the various cell lines, with higher sensitivity to cytotoxicity by the cells with higher proliferation rate and reduced doubling time (human keratinocytes). Moreover, to exclude an inflammatory effect at the not cytotoxic WD concentrations, the expression of the main inducible markers of inflammation, cyclooxygenase-2 (COX-2) and microsomal prostaglandin E synthase-1 (mPGES-1), were assessed, and no improvement was found both after brief and prolonged exposure. In conclusion, our data exclude any inflammatory and cytotoxic effect at the lowest WD concentrations, namely 0.07% and 0.04%, mimicking some recommended dilutions of the product and the potential exposure doses for the operators in agriculture. Nevertheless, higher concentrations showed a safe profile for short time usage, but caution should be used by farmers following persistent product exposure.
Collapse
|
7
|
Martelli A, Piragine E, Gorica E, Citi V, Testai L, Pagnotta E, Lazzeri L, Pecchioni N, Ciccone V, Montanaro R, Di Cesare Mannelli L, Ghelardini C, Brancaleone V, Morbidelli L, Calderone V. The H 2S-Donor Erucin Exhibits Protective Effects against Vascular Inflammation in Human Endothelial and Smooth Muscle Cells. Antioxidants (Basel) 2021; 10:antiox10060961. [PMID: 34203803 PMCID: PMC8232611 DOI: 10.3390/antiox10060961] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023] Open
Abstract
Preservation of vascular wall integrity against degenerative processes associated with ageing, fat-rich diet and metabolic diseases is a timely therapeutical challenge. The loss of endothelial function and integrity leads to cardiovascular diseases and multiorgan inflammation. The protective effects of the H2S-donor erucin, an isothiocyanate purified by Eruca sativa Mill. seeds, were evaluated on human endothelial and vascular smooth muscle cells. In particular, erucin actions were evaluated on cell viability, ROS, caspase 3/7, inflammatory markers levels and the endothelial hyperpermeability in an inflammatory model associated with high glucose concentrations (25 mM, HG). Erucin significantly prevented the HG-induced decrease in cell viability as well as the increase in ROS, caspase 3/7 activation, and TNF-α and IL-6 levels. Similarly, erucin suppressed COX-2 and NF-κB upregulation associated with HG exposure. Erucin also caused a significant inhibition of p22phox subunit expression in endothelial cells. In addition, erucin significantly prevented the HG-induced increase in endothelial permeability as also confirmed by the quantification of the specific markers VE-Cadherin and ZO-1. In conclusion, our results assess anti-inflammatory and antioxidant effects by erucin in vascular cells undergoing HG-induced inflammation and this protection parallels the preservation of endothelial barrier properties.
Collapse
Affiliation(s)
- Alma Martelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.P.); (E.G.); (V.C.); (L.T.)
- Interdepartmental Research Center “Nutrafood: Nutraceutica e Alimentazione per la Salute”, University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Center “Biology and Pathology of Ageing”, University of Pisa, 56126 Pisa, Italy
- Correspondence: (A.M.); (V.C.)
| | - Eugenia Piragine
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.P.); (E.G.); (V.C.); (L.T.)
| | - Era Gorica
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.P.); (E.G.); (V.C.); (L.T.)
| | - Valentina Citi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.P.); (E.G.); (V.C.); (L.T.)
| | - Lara Testai
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.P.); (E.G.); (V.C.); (L.T.)
- Interdepartmental Research Center “Nutrafood: Nutraceutica e Alimentazione per la Salute”, University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Center “Biology and Pathology of Ageing”, University of Pisa, 56126 Pisa, Italy
| | - Eleonora Pagnotta
- Research Centre for Cereal and Industrial Crops, CREA Council for Agricultural Research and Economics, Via di Corticella 128, 40134 Bologna, Italy; (E.P.); (L.L.)
| | - Luca Lazzeri
- Research Centre for Cereal and Industrial Crops, CREA Council for Agricultural Research and Economics, Via di Corticella 128, 40134 Bologna, Italy; (E.P.); (L.L.)
| | - Nicola Pecchioni
- Research Centre for Cereal and Industrial Crops, CREA Council for Agricultural Research and Economics, S.S. 673 Km 25,200, 71122 Foggia, Italy;
| | - Valerio Ciccone
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy; (V.C.); (L.M.)
| | - Rosangela Montanaro
- Department of Science, University of Basilicata, Via dell’Ateneo lucano, 10, 85100 Potenza, Italy; (R.M.); (V.B.)
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health–NEUROFARBA–Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy; (L.D.C.M.); (C.G.)
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health–NEUROFARBA–Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy; (L.D.C.M.); (C.G.)
| | - Vincenzo Brancaleone
- Department of Science, University of Basilicata, Via dell’Ateneo lucano, 10, 85100 Potenza, Italy; (R.M.); (V.B.)
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy; (V.C.); (L.M.)
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.P.); (E.G.); (V.C.); (L.T.)
- Interdepartmental Research Center “Nutrafood: Nutraceutica e Alimentazione per la Salute”, University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Center “Biology and Pathology of Ageing”, University of Pisa, 56126 Pisa, Italy
- Correspondence: (A.M.); (V.C.)
| |
Collapse
|
8
|
Effect of NIR Laser Therapy by MLS-MiS Source on Fibroblast Activation by Inflammatory Cytokines in Relation to Wound Healing. Biomedicines 2021; 9:biomedicines9030307. [PMID: 33809724 PMCID: PMC8002295 DOI: 10.3390/biomedicines9030307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/05/2021] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
The fine control of inflammation following injury avoids fibrotic scars or impaired wounds. Due to side effects by anti-inflammatory drugs, the research is continuously active to define alternative therapies. Among them, physical countermeasures such as photobiomodulation therapy (PBMT) are considered effective and safe. To study the cellular and molecular events associated with the anti-inflammatory activity of PBMT by a dual-wavelength NIR laser source, human dermal fibroblasts were exposed to a mix of inflammatory cytokines (IL-1β and TNF-α) followed by laser treatment once a day for three days. Inducible inflammatory key enzymatic pathways, as iNOS and COX-2/mPGES-1/PGE2, were upregulated by the cytokine mix while PBMT reverted their levels and activities. The same behavior was observed with the proangiogenic factor vascular endothelial growth factor (VEGF), involved in neovascularization of granulation tissue. From a molecular point of view, PBMT retained NF-kB cytoplasmatic localization. According to a change in cell morphology, differences in expression and distribution of fundamental cytoskeletal proteins were observed following treatments. Tubulin, F-actin, and α-SMA changed their organization upon cytokine stimulation, while PBMT reestablished the basal localization. Cytoskeletal rearrangements occurring after inflammatory stimuli were correlated with reorganization of membrane α5β1 and fibronectin network as well as with their upregulation, while PBMT induced significant downregulation. Similar changes were observed for collagen I and the gelatinolytic enzyme MMP-1. In conclusion, the present study demonstrates that the proposed NIR laser therapy is effective in controlling fibroblast activation induced by IL-1β and TNF-α, likely responsible for a deleterious effect of persistent inflammation.
Collapse
|
9
|
Ristori E, Cicaloni V, Salvini L, Tinti L, Tinti C, Simons M, Corti F, Donnini S, Ziche M. Amyloid-β Precursor Protein APP Down-Regulation Alters Actin Cytoskeleton-Interacting Proteins in Endothelial Cells. Cells 2020; 9:cells9112506. [PMID: 33228083 PMCID: PMC7699411 DOI: 10.3390/cells9112506] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 11/16/2022] Open
Abstract
The amyloid-β precursor protein (APP) is a ubiquitous membrane protein often associated with Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). Despite its role in the development of the pathogenesis, APP exerts several physiological roles that have been mainly investigated in neuronal tissue. To date, the role of APP in vasculature and endothelial cells has not been fully elucidated. In this study, we used molecular and proteomic approaches to identify and investigate major cellular targets of APP down-regulation in endothelial cells. We found that APP is necessary for endothelial cells proliferation, migration and adhesion. The loss of APP alters focal adhesion stability and cell-cell junctions' expression. Moreover, APP is necessary to mediate endothelial response to the VEGF-A growth factor. Finally, we document that APP propagates exogenous stimuli and mediates cellular response in endothelial cells by modulating the Scr/FAK signaling pathway. Thus, the intact expression and processing of APP is required for normal endothelial function. The identification of molecular mechanisms responsible for vasoprotective properties of endothelial APP may have an impact on clinical efforts to preserve and protect healthy vasculature in patients at risk of the development of cerebrovascular disease and dementia including AD and CAA.
Collapse
Affiliation(s)
- Emma Ristori
- Department of Life Science, University of Siena, 53100 Siena, Italy;
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Vittoria Cicaloni
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Laura Salvini
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Laura Tinti
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Cristina Tinti
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Michael Simons
- Yale Cardiovascular Research Center, 300 George Street, New Haven, CT 06511, USA; (M.S.); (F.C.)
- Departments of Medicine (Cardiology) and Cell Biology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Federico Corti
- Yale Cardiovascular Research Center, 300 George Street, New Haven, CT 06511, USA; (M.S.); (F.C.)
| | - Sandra Donnini
- Department of Life Science, University of Siena, 53100 Siena, Italy;
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
- Correspondence: (S.D.); (M.Z.); Tel.: +39-0577-235382 (S.D.)
| | - Marina Ziche
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- Correspondence: (S.D.); (M.Z.); Tel.: +39-0577-235382 (S.D.)
| |
Collapse
|
10
|
Pharmacological Inhibition of CA-IX Impairs Tumor Cell Proliferation, Migration and Invasiveness. Int J Mol Sci 2020; 21:ijms21082983. [PMID: 32340282 PMCID: PMC7215745 DOI: 10.3390/ijms21082983] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 12/25/2022] Open
Abstract
Carbonic anhydrase IX (CA-IX) plays a pivotal role in regulation of pH in tumor milieu catalyzing carbonic acid formation by hydrating CO2. An acidification of tumor microenvironment contributes to tumor progression via multiple processes, including reduced cell-cell adhesion, increased migration and matrix invasion. We aimed to assess whether the pharmacological inhibition of CA-IX could impair tumor cell proliferation and invasion. Tumor epithelial cells from breast (MDA-MB-231) and lung (A549) cancer were used to evaluate the cytotoxic effect of sulfonamide CA-IX inhibitors. Two CA-IX enzyme blockers were tested, SLC-0111 (at present in phase Ib clinical trial) and AA-06-05. In these cells, the drugs inhibited cell proliferation, migration and invasion through shifting of the mesenchymal phenotype toward an epithelial one and by impairing matrix metalloprotease-2 (MMP-2) activity. The antitumor activity was elicited via apoptosis pathway activation. An upregulation of p53 was observed, which in turn regulated the activation of caspase-3. Inhibition of proteolytic activity was accompanied by upregulation of the endogenous tissue inhibitor TIMP-2. Collectively, these data confirm the potential use of CA-IX inhibitors, and in particular SLC-0111 and AA-06-05, as agents to be further developed, alone or in combination with other conventional anticancer drugs.
Collapse
|
11
|
Terzuoli E, Costanza F, Ciccone V, Ziche M, Morbidelli L, Donnini S. mPGES-1 as a new target to overcome acquired resistance to gefitinib in non-small cell lung cancer cell lines. Prostaglandins Other Lipid Mediat 2019; 143:106344. [PMID: 31207300 DOI: 10.1016/j.prostaglandins.2019.106344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/18/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023]
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) as gefitinib are standard treatment of non-small cell lung cancer (NSCLC), but resistance often occurs. This study demonstrates that NSCLC cells resistant to gefitinib (GR cells) displayed a significantly higher microsomal prostaglandin E synthase-1 (mPGES-1) expression and activity than parental cells. Overexpression of mPGES-1/prostaglandin E-2 (PGE-2) signaling in GR cells was associated with acquisition of mesenchymal and stem-like cell properties, nuclear EGFR translocation and tolerance to cisplatin. mPGES-1 inhibition reduced mesenchymal and stem-like properties, and nuclear EGFR translocation in GR cells. Consistently, inhibition of mPGES-1 activity enhanced sensitivity to cisplatin and responsiveness to gefitinib in GR cells. We propose the mPGES-1/PGE-2 signaling as a potential target for treating aggressive and resistant lung cancers.
Collapse
Affiliation(s)
- Erika Terzuoli
- Department of Medical and Surgical Sciences and Neurosciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| | - Filomena Costanza
- Department of Life Sciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| | - Valerio Ciccone
- Department of Life Sciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| | - Marina Ziche
- Department of Medical and Surgical Sciences and Neurosciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| |
Collapse
|
12
|
Ciccone V, Terzuoli E, Donnini S, Giachetti A, Morbidelli L, Ziche M. Stemness marker ALDH1A1 promotes tumor angiogenesis via retinoic acid/HIF-1α/VEGF signalling in MCF-7 breast cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:311. [PMID: 30541574 PMCID: PMC6291966 DOI: 10.1186/s13046-018-0975-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/21/2018] [Indexed: 12/12/2022]
Abstract
Background Aldehyde dehydrogenase 1A1 (ALDH1A1), a member of aldehyde dehydrogenase family, is a marker of stemness in breast cancer. During tumor progression cancer stem cells (CSCs) have been reported to secrete angiogenic factors to orchestrate the formation of pathological angiogenesis. This vasculature can represent the source of self-renewal of CSCs and the route for further tumor spreading. The aim of the present study has been to assess whether ALDH1A1 controls the output of angiogenic factors in breast cancer cells and regulates tumor angiogenesis in a panel of in vitro and in vivo models. Methods Stemness status of breast cancer cells was evaluated by the ability to form turmorspheres in vitro. A transwell system was used to assess the angiogenic features of human umbilical vein endothelial cells (HUVEC) when co-cultured with breast cancer cells MCF-7 harboring different levels of ALDH1A1. Under these conditions, we survey endothelial proliferation, migration, tube formation and permeability. Moreover, in vivo, MCF-7 xenografts in immunodeficient mice allow to evaluate blood flow, expression of angiogenic factors and microvascular density (MVD). Results In MCF-7 we observed that ALDH1A1 activity conferred stemness property and its expression correlated with an activation of angiogenic factors. In particular we observed a significant upregulation of hypoxia inducible factor-1α (HIF-1α) and proangiogenic factors, such as vascular endothelial growth factor (VEGF). High levels of ALDH1A1, through the retinoic acid pathway, were significantly associated with VEGF-mediated angiogenesis in vitro. Co-culture of HUVEC with ALDH1A1 expressing tumor cells promoted endothelial proliferation, migration, tube formation and permeability. Conversely, downregulation of ALDH1A1 in MCF-7 resulted in reduction of proangiogenic factor release/expression and impaired HUVEC angiogenic functions. In vivo, when subcutaneously implanted in immunodeficient mice, ALDH1A1 overexpressing breast tumor cells displayed a higher expression of VEGF and MVD. Conclusion In breast tumors, ALDH1A1 expression primes a permissive microenvironment by promoting tumor angiogenesis via retinoic acid dependent mechanism. In conclusion, ALDH1A1 might be associated to progression and diffusion of breast cancer. Electronic supplementary material The online version of this article (10.1186/s13046-018-0975-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Valerio Ciccone
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Erika Terzuoli
- Department of Medicine, Surgery and Neuroscience, University of Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Antonio Giachetti
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100, Siena, Italy.
| | - Marina Ziche
- Department of Medicine, Surgery and Neuroscience, University of Siena, Via A. Moro 2, 53100, Siena, Italy.
| |
Collapse
|
13
|
ALDH2 Activity Reduces Mitochondrial Oxygen Reserve Capacity in Endothelial Cells and Induces Senescence Properties. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9765027. [PMID: 30538807 PMCID: PMC6261243 DOI: 10.1155/2018/9765027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/31/2018] [Accepted: 09/09/2018] [Indexed: 12/18/2022]
Abstract
Endothelial cells (ECs) are dynamic cells that turn from growth into senescence, the latter being associated with cellular dysfunction, altered metabolism, and age-related cardiovascular diseases. Aldehyde dehydrogenase 2 (ALDH2) is a mitochondrial enzyme metabolizing acetaldehyde and other toxic aldehydes, such as 4-hydroxynonenal (4-HNE). In conditions in which lipid peroxidation products and reactive oxygen species (ROS) are accumulated, ECs become dysfunctional and significantly contribute to the progression of vascular-dependent diseases. The aim of the present study has been to investigate whether inhibition of ALDH2 alters endothelial functions together with the impairment of bioenergetic functions, accelerating the acquisition of a senescent phenotype. HUVECs transfected with siRNA targeting ALDH2 or treated with daidzin, an ALDH2 inhibitor, were used in this study. We observed an alteration in cell morphology associated with endothelial dysfunctions. Loss of ALDH2 reduced cell proliferation and migration and increased paracellular permeability. To assess bioenergetic function in intact ECs, extracellular flux analysis was carried out to establish oxygen consumption rates (OCR). We observed a decrease in mitochondrial respiration and reserve capacity that coincided with SA-β-Gal accumulation and an increase in p21 and p53 expression in siALDH2 or daidzin-treated HUVECs. Treatment with N-acetyl-L-cysteine (NAC) reduced endothelial dysfunctions mediated by siALDH2, indicating that oxidative stress downstream to siALDH2 plays an instrumental role. Our results highlight that ALDH2 impairment accelerates the acquisition of a premature senescent phenotype, a change likely to be associated with the observed reduction of mitochondrial respiration and reserve capacity.
Collapse
|
14
|
Cross-talk between endogenous H 2S and NO accounts for vascular protective activity of the metal-nonoate Zn(PipNONO)Cl. Biochem Pharmacol 2018; 152:143-152. [PMID: 29588193 DOI: 10.1016/j.bcp.2018.03.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 03/22/2018] [Indexed: 02/08/2023]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are now recognized as gaseous transmitters with many cardiovascular protective properties. The present study concerns the possibility that NO donors can also function through endogenous activation of NO and H2S pathways. Based on the previous characterization of a novel metal-nonoate, Ni(PipNONO)Cl, our aim was: 1) to study the effects of a zinc based compound, Zn(PipNONO)Cl, on vascular endothelial and smooth muscle cells, and 2) to assess the role and interplay between endogenous NO and H2S promoted by the nonoate. Zn(PipNONO)Cl completely reproduced the vasodilation elicited by Ni(PipNONO)Cl. In the presence of endothelium, preincubation with Zn(PipNONO)Cl sensitized the intima to acetylcholine-induced vasodilation. When tested on cultured endothelial cells, Zn(PipNONO)Cl prompted PI-3K/Akt- and MAPK/ERK1/2-mediated survival. Nitrite levels indicated fast NO release (due to the molecule) and delayed (1-6 h) NO production linked to PI-3K/Akt-dependent eNOS activation. In the same time frame (1-6 h), significant CSE-dependent H2S levels were detected in response to Zn(PipNONO)Cl. The mechanisms responsible for H2S increase seemed to depend on the NONO moiety/sGC/cGMP pathway and zinc-associated ROS production. Our results indicate that endogenous H2S and NO were produced after fast NO release from Zn(PipNONO)Cl, contributing to the vascular endothelium protective effect. The effect was partially reproduced on smooth muscle cells, where Zn(PipNONO)Cl inhibited cell proliferation and migration. In conclusion, vasorelaxant effects, with complementary activities on endothelium and smooth muscle cells, are elicited by the novel metal-nonoate Zn(PipNONO)Cl.
Collapse
|
15
|
Ciccone V, Monti M, Monzani E, Casella L, Morbidelli L. The metal-nonoate Ni(SalPipNONO) inhibits in vitro tumor growth, invasiveness and angiogenesis. Oncotarget 2018; 9:13353-13365. [PMID: 29568362 PMCID: PMC5862583 DOI: 10.18632/oncotarget.24350] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/25/2018] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) exerts conflicting effect on tumor growth and progression, depending on its concentration. We aimed to characterize the anti-cancer activity of a new NO donor, Ni(SalPipNONO) belonging to the class of metal-nonoates, in epithelial derived tumor cells, finally exploring its antiangiogenic properties. Tumor epithelial cells were screened to evaluate the cytotoxic effect of Ni(SalPipNONO), which was able to inhibit cell proliferation in a dose dependent manner, being more effective than the commercial DETA/NO. The human lung carcinoma cells A549 were chosen as model to study the anti-cancer mechanisms exerted by the compound. In these cells, Ni(SalPipNONO) inhibited clonogenicity and cell invasion, while promoting apoptosis. The antitumor activity was partly due to NO-cGMP dependent pathway, contributing to reduced cell number and apoptosis, and partly to the salicylaldehyde moiety and reactive oxygen species (ROS) activated ERK1/2 signaling converging on p53 dependent caspase-3 cleavage. An additional contribution by downstream cycloxygenase-2 (COX-2) derived cyclopentenones may explain the tumor inhibitory activities. As NO has been described to affect tumor angiogenesis, we checked this activity both on tumor and endothelial cell co-cultures and in Matrigel in vivo assay. Our data document that Ni(SalPipNONO) was able to both reduce angiogenic factor expression by tumor cells acting on hypoxia inducible factor-1α (HIF-1 α) level, and endothelial cell functions related to angiogenesis. Collectively, these data confirm the potential use of NO donors and in particular Ni(SalPipNONO) acting through multiple mechanisms, as an agent to be further developed to be used alone or in combination with conventional therapy.
Collapse
Affiliation(s)
- Valerio Ciccone
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Martina Monti
- Department of Molecular Medicine and Development, University of Siena, Siena, Italy.,Noxamet Ltd, Milan, Italy
| | - Enrico Monzani
- Noxamet Ltd, Milan, Italy.,Department of Chemistry, University of Pavia, Pavia, Italy
| | - Luigi Casella
- Noxamet Ltd, Milan, Italy.,Department of Chemistry, University of Pavia, Pavia, Italy
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Siena, Italy.,Noxamet Ltd, Milan, Italy
| |
Collapse
|
16
|
Blei F. Update March 2017. Lymphat Res Biol 2017. [DOI: 10.1089/lrb.2017.29019.fb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|