1
|
Jyotirmaya SS, Rath S, Dandapat J. Redox imbalance driven epigenetic reprogramming and cardiovascular dysfunctions: phytocompounds for prospective epidrugs. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156380. [PMID: 39827814 DOI: 10.1016/j.phymed.2025.156380] [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: 03/12/2024] [Revised: 05/10/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are the major contributor to global mortality and are gaining incremental attention following the COVID-19 outbreak. Epigenetic events such as DNA methylation, histone modifications, and non-coding RNAs have a significant impact on the incidence and onset of CVDs. Altered redox status is one of the major causative factors that regulate epigenetic pathways linked to CVDs. Various bioactive phytocompounds used in alternative therapies including Traditional Chinese Medicines (TCM) regulate redox balance and epigenetic phenomena linked to CVDs. Phytocompound-based medications are in the limelight for the development of cost-effective drugs with the least side effects, which will have immense therapeutic applications. PURPOSE This review comprehends certain risk factors associated with CVDs and triggered by oxidative stress-driven epigenetic remodelling. Further, it critically evaluates the pharmacological efficacy of phytocompounds as inhibitors of HAT/HDAC and DNMTs as well as miRNAs regulator that lowers the incidence of CVDs, aiming for new candidates as prospective epidrugs. METHODS PRISMA flow approach has been adopted for systematic literature review. Different Journals, computational databases, search engines such as Google Scholar, PubMed, Science Direct, Scopus, and ResearchGate were used to collect online information for literature survey. Statistical information collected from the World Health Organization (WHO) site (https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds)) and the American Heart Association of Heart Disease and Stroke reported the international and national status of CVDs. RESULTS The meta-analysis of various studies is elucidated in the literature, shedding light on major risk factors such as socioeconomic parameters, which contribute highly to redox imbalance, epigenetic modulations, and CVDs. Going forward, redox imbalance driven epigenetic regulations include changes in DNA methylation status, histone modifications and non-coding RNAs expression pattern which further regulates global as well as promoter modification of various transcription factors leading to the onset of CVDs. Further, the role of various bioactive compounds used in herbal medicine, including TCM for redox regulation and epigenetic modifications are discussed. Pharmacological safety doses and different phases of clinical trials of these phytocompounds are elaborated on, which shed light on the acceptance of these phytocompounds as prospective drugs. CONCLUSION This review suggests a strong linkage between therapeutic and preventive measures against CVDs by targeting redox imbalance-driven epigenetic reprogramming using phytocompounds as prospective epidrugs. Future in-depth research is required to evaluate the possible molecular mechanisms behind the phytocompound-mediated epigenetic reprogramming and oxidative stress management during CVD progression.
Collapse
Affiliation(s)
| | - Suvasmita Rath
- Post-graduate Department of Biotechnology, Utkal University, Bhubaneswar, 751004, Odisha, India.; Centre of Environment, Climate Change and Public Health, Utkal University, Vani Vihar, Bhubaneswar,751004, Odisha, India
| | - Jagneshwar Dandapat
- Post-graduate Department of Biotechnology, Utkal University, Bhubaneswar, 751004, Odisha, India.; Centre of Excellence in Integrated Omics and Computational Biology, Utkal University, Bhubaneswar 751004, Odisha, India..
| |
Collapse
|
2
|
Alam MI, Sami N, Alam A, Wazib S, Dhyani N, Afghan S, Ansari MA. Estrogen-mediated modulation of sterile inflammatory markers and baroreflex sensitivity in ovariectomized female Wistar rats. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e230521. [PMID: 39876967 PMCID: PMC11771758 DOI: 10.20945/2359-4292-2023-0521] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 07/16/2024] [Indexed: 01/31/2025]
Abstract
Objective This study aims to explore the role of estrogen in providing cardioprotective benefits to premenopausal women, examining how hormonal differences between sexes influence the prevalence of cardiovascular diseases (CVDs) in women. Materials and methods Eighteen female Wistar rats were equally distributed into three treatment groups. Animals in Group I (sham-operated) and Group II (ovariectomized [OVX]) received oral saline solution at a dose of 2 mL/kg. Group III (OVX+E2) received oral E2 2 µg/mL/kg after ovariectomy. Hemodynamic parameters and baroreflex sensitivity were determined in all groups. Plasma levels of malondialdehyde (MDA), superoxide dismutase (SOD), and nitric oxide (NO) were measured, along with those of the inflammatory markers tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and high mobility group box-1 (HMGB-1). Results The OVX group, compared with the sham-operated group, displayed significantly altered hemodynamic parameters and baroreflex sensitivity, along with elevated MDA levels and decreased SOD and NO levels. This group also had higher levels of inflammatory cytokines than the sham-operated group. In the absence of estrogen, these factors led to the advancement of cardiovascular abnormalities. In the OVX+E2 group, estrogen treatment considerably improved baroreflex sensitivity and hemodynamic profile while reducing the expression of inflammatory cytokines compared with the OVX group, demonstrating anti-inflammatory actions of estrogen. Conclusion Estrogen mediates cardioprotection by improving baroreflex sensitivity in ovariectomized Wistar rats through modulation of the NO pathway.
Collapse
Affiliation(s)
- Md. Iqbal Alam
- Jamia Hamdard UniversityHamdard Institute of Medical Sciences and ResearchDepartment of PhysiologyNew DelhiIndiaDepartment of Physiology, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard University, New Delhi
| | - Naba Sami
- Jamia Hamdard UniversityHamdard Institute of Medical Sciences and ResearchDepartment of PhysiologyNew DelhiIndiaDepartment of Physiology, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard University, New Delhi
| | - Aftab Alam
- Coventry UniversitySchool of Life SciencesCoventryUKSchool of Life Sciences, Coventry University, Coventry, UK
| | - Sheema Wazib
- Jamia Hamdard UniversityHamdard Institute of Medical Sciences and ResearchDepartment of PhysiologyNew DelhiIndiaDepartment of Physiology, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard University, New Delhi
| | - Neha Dhyani
- Jamia Hamdard UniversityHamdard Institute of Medical Sciences and ResearchDepartment of PhysiologyNew DelhiIndiaDepartment of Physiology, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard University, New Delhi
| | - Sher Afghan
- Jamia Hamdard UniversityHamdard Institute of Medical Sciences and ResearchDepartment of PhysiologyNew DelhiIndiaDepartment of Physiology, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard University, New Delhi
| | - Mairaj Ahmed Ansari
- Jamia HamdardSchool of Chemical & Life SciencesDepartment of BiotechnologyNew DelhiIndiaDepartment of Biotechnology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi
| |
Collapse
|
3
|
Lewis A. A hypothesis of teleological evolution, via endogenous acetylcholine, nitric oxide, and calmodulin pathways. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 188:68-76. [PMID: 38552848 DOI: 10.1016/j.pbiomolbio.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/30/2024] [Accepted: 03/22/2024] [Indexed: 04/08/2024]
Abstract
The Extended Evolutionary Synthesis (EES) addresses the issues in evolutionary biology which cannot be explained by neo-Darwinian theory. The EES paradigm recognises teleology and agency in living systems, and identifies that organisms can directly affect their evolutionary trajectory in a goal-directed manner, yet the physiological pathways via which this occurs remain unidentified. Here, I propose a physiological pathway via which organisms can alter their genotype and phenotype by making behavioural decisions with respect their activity levels, partitioning of resources either toward growth, defence against disease, or their behavioural response to stressors. Specifically, I hypothesize that agential, teleological decisions mediated by acetylcholine result in induced nitric oxide (NO) activity, which regulates metabolism, blood flow, and immune response. Nitric oxide, however, is also a key epigenetic molecule, being involved in DNA acetylation, methylation, and de-methylation. Further, NO alters the histone complexes which scaffold nuclear DNA strands, and is thus a good candidate in identifying a system which allows an organisms to make teleological genetic changes. The proposed mechanisms of inheritance of these genetic changes is via the paternal line, whereby epigenetic changes in the somatic Sertoli cells in animals are transcribed by mRNA and included in the germline cells - the male gametes. The microsporangium in plants, and the sporophore cells in fungi, meanwhile, are proposed to form similar systems in response to sensory detection of stressors. Whilst the hypothesis is presented as a simplified model for future testing, it opens new avenues for study in evolutionary biology.
Collapse
|
4
|
Frühauf A, Behringer M, Meyer-Almes FJ. Significance of Five-Membered Heterocycles in Human Histone Deacetylase Inhibitors. Molecules 2023; 28:5686. [PMID: 37570656 PMCID: PMC10419652 DOI: 10.3390/molecules28155686] [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: 05/29/2023] [Revised: 07/15/2023] [Accepted: 07/15/2023] [Indexed: 08/13/2023] Open
Abstract
Five-membered heteroaromatic rings, in particular, have gained prominence in medicinal chemistry as they offer enhanced metabolic stability, solubility and bioavailability, crucial factors in developing effective drugs. The unique physicochemical properties and biological effects of five-membered heterocycles have positioned them as key structural motifs in numerous clinically effective drugs. Hence, the exploration of five-ring heterocycles remains an important research area in medicinal chemistry, with the aim of discovering new therapeutic agents for various diseases. This review addresses the incorporation of heteroatoms such as nitrogen, oxygen and sulfur into the aromatic ring of these heterocyclic compounds, enhancing their polarity and facilitating both aromatic stacking interactions and the formation of hydrogen bonds. Histone deacetylases are present in numerous multiprotein complexes within the epigenetic machinery and play a central role in various cellular processes. They have emerged as important targets for cancer, neurodegenerative diseases and other therapeutic indications. In histone deacetylase inhibitors (HDACi's), five-ring heterocycles perform various functions as a zinc-binding group, a linker or head group, contributing to binding activity and selective recognition. This review focuses on providing an up-to-date overview of the different five-membered heterocycles utilized in HDACi motifs, highlighting their biological properties. It summarizes relevant publications from the past decade, offering insights into the recent advancements in this field of research.
Collapse
Affiliation(s)
- Anton Frühauf
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences, Haardtring 100, 64295 Darmstadt, Germany
| | - Martin Behringer
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences, Haardtring 100, 64295 Darmstadt, Germany
| | - Franz-Josef Meyer-Almes
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences, Haardtring 100, 64295 Darmstadt, Germany
| |
Collapse
|
5
|
Salvatori L, Malatesta S, Illi B, Somma MP, Fionda C, Stabile H, Fontanella RA, Gaetano C. Nitric Oxide Prevents Glioblastoma Stem Cells' Expansion and Induces Temozolomide Sensitization. Int J Mol Sci 2023; 24:11286. [PMID: 37511047 PMCID: PMC10379318 DOI: 10.3390/ijms241411286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Glioblastoma multiforme (GBM) has high mortality and recurrence rates. Malignancy resilience is ascribed to Glioblastoma Stem Cells (GSCs), which are resistant to Temozolomide (TMZ), the gold standard for GBM post-surgical treatment. However, Nitric Oxide (NO) has demonstrated anti-cancer efficacy in GBM cells, but its potential impact on GSCs remains unexplored. Accordingly, we investigated the effects of NO, both alone and in combination with TMZ, on patient-derived GSCs. Experimentally selected concentrations of diethylenetriamine/NO adduct and TMZ were used through a time course up to 21 days of treatment, to evaluate GSC proliferation and death, functional recovery, and apoptosis. Immunofluorescence and Western blot analyses revealed treatment-induced effects in cell cycle and DNA damage occurrence and repair. Our results showed that NO impairs self-renewal, disrupts cell-cycle progression, and expands the quiescent cells' population. Consistently, NO triggered a significant but tolerated level of DNA damage, but not apoptosis. Interestingly, NO/TMZ cotreatment further inhibited cell cycle progression, augmented G0 cells, induced cell death, but also enhanced DNA damage repair activity. These findings suggest that, although NO administration does not eliminate GSCs, it stunts their proliferation, and makes cells susceptible to TMZ. The resulting cytostatic effect may potentially allow long-term control over the GSCs' subpopulation.
Collapse
Affiliation(s)
- Luisa Salvatori
- Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Sapienza University of Rome, 00185 Rome, Italy
| | - Silvia Malatesta
- Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Sapienza University of Rome, 00185 Rome, Italy
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy
| | - Barbara Illi
- Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Patrizia Somma
- Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Sapienza University of Rome, 00185 Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Helena Stabile
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Rosaria Anna Fontanella
- Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Sapienza University of Rome, 00185 Rome, Italy
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Carlo Gaetano
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| |
Collapse
|
6
|
Omidkhah N, Ghodsi R. NO-HDAC dual inhibitors. Eur J Med Chem 2021; 227:113934. [PMID: 34700268 DOI: 10.1016/j.ejmech.2021.113934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/08/2021] [Accepted: 10/17/2021] [Indexed: 12/11/2022]
Abstract
HDAC inhibitors and NO donors have both demonstrated independently broad therapeutic potential in a variety of diseases. Borretto et al. presented the topic of NO-HDAC dual inhibitors for the first time in 2013 as an attractive new topic. Here we collected the general structure of all synthesized NO-HDAC dual inhibitors, lead compounds, synthesis methods and biological features of the most potent dual NO-HDAC inhibitor in each category with the intention of assisting in the synthesis and optimization of new drug-like compounds for diverse diseases. Based on studies done so far, NO-HDAC dual inhibitors have displayed satisfactory results against wound healing (3), heart hypertrophy (3), inflammatory, cardiovascular, neuromuscular illnesses (11a-11e) and cancer (6a-6o, 9a-9d, 10a-10d, 16 and 17). NO-HDAC dual inhibitors can have high therapeutic potential for various diseases due to their new properties, NO properties, HDAC inhibitor properties and also due to the effects of NO on HDAC enzymes.
Collapse
Affiliation(s)
- Negar Omidkhah
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Razieh Ghodsi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Guo Z, Mo Z. Regulation of endothelial cell differentiation in embryonic vascular development and its therapeutic potential in cardiovascular diseases. Life Sci 2021; 276:119406. [PMID: 33785330 DOI: 10.1016/j.lfs.2021.119406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/05/2021] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
During vertebrate development, the cardiovascular system begins operating earlier than any other organ in the embryo. Endothelial cell (EC) forms the inner lining of blood vessels, and its extensive proliferation and migration are requisite for vasculogenesis and angiogenesis. Many aspects of cellular biology are involved in vasculogenesis and angiogenesis, including the tip versus stalk cell specification. Recently, epigenetics has attracted growing attention in regulating embryonic vascular development and controlling EC differentiation. Some proteins that regulate chromatin structure have been shown to be directly implicated in human cardiovascular diseases. Additionally, the roles of important EC signaling such as vascular endothelial growth factor and its receptors, angiopoietin-1 and tyrosine kinase containing immunoglobulin and epidermal growth factor homology domain-2, and transforming growth factor-β in EC differentiation during embryonic vasculature development are briefly discussed in this review. Recently, the transplantation of human induced pluripotent stem cell (iPSC)-ECs are promising approaches for the treatment of ischemic cardiovascular disease including myocardial infarction. Patient-specific iPSC-derived EC is a potential new target to study differences in gene expression or response to drugs. However, clinical application of the iPSC-ECs in regenerative medicine is often limited by the challenges of maintaining cell viability and function. Therefore, novel insights into the molecular mechanisms underlying EC differentiation might provide a better understanding of embryonic vascular development and bring out more effective EC-based therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Zi Guo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaohui Mo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
8
|
Oxidative, Reductive, and Nitrosative Stress Effects on Epigenetics and on Posttranslational Modification of Enzymes in Cardiometabolic Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8819719. [PMID: 33204398 PMCID: PMC7649698 DOI: 10.1155/2020/8819719] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/09/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023]
Abstract
Oxidative (OS), reductive (RS), and nitrosative (NSS) stresses produce carbonylation, glycation, glutathionylation, sulfhydration, nitration, and nitrosylation reactions. OS, RS, and NSS are interrelated since RS results from an overactivation of antioxidant systems and NSS is the result of the overactivation of the oxidation of nitric oxide (NO). Here, we discuss the general characteristics of the three types of stress and the way by which the reactions they induce (a) damage the DNA structure causing strand breaks or inducing the formation of 8-oxo-d guanosine; (b) modify histones; (c) modify the activities of the enzymes that determine the establishment of epigenetic cues such as DNA methyl transferases, histone methyl transferases, acetyltransferases, and deacetylases; (d) alter DNA reparation enzymes by posttranslational mechanisms; and (e) regulate the activities of intracellular enzymes participating in metabolic reactions and in signaling pathways through posttranslational modifications. Furthermore, the three types of stress may establish new epigenetic marks through these reactions. The development of cardiometabolic disorders in adult life may be programed since early stages of development by epigenetic cues which may be established or modified by OS, RS, and NSS. Therefore, the three types of stress participate importantly in mediating the impact of the early life environment on later health and heritability. Here, we discuss their impact on cardiometabolic diseases. The epigenetic modifications induced by these stresses depend on union and release of chemical residues on a DNA sequence and/or on amino acid residues in proteins, and therefore, they are reversible and potentially treatable.
Collapse
|
9
|
Geib T, Iacob C, Jribi R, Fernandes J, Benderdour M, Sleno L. Identification of 4-hydroxynonenal-modified proteins in human osteoarthritic chondrocytes. J Proteomics 2020; 232:104024. [PMID: 33122130 DOI: 10.1016/j.jprot.2020.104024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 10/06/2020] [Accepted: 10/22/2020] [Indexed: 11/19/2022]
Abstract
The α,β-unsaturated aldehyde 4-hydroxynonenal (HNE) is formed through lipid peroxidation during oxidative stress. As a highly reactive electrophile, it is able to form adducts with various biomolecules, including proteins. These protein modifications could modulate many signaling pathways, as well as cell differentiation and proliferation, and thus could be highly important in the context of the extracellular matrix and degradation of articular cartilage. This study specifically investigated the role of HNE as a bioactive molecule in chondrocytes of osteoarthritis (OA) patients. Chondrocyte extracts of OA and non-OA patients were analyzed for HNE binding using Western blot and bottom-up LC-MS/MS analyses. HNE-modified histones, H2A and H2B, and histone deacetylase were identified using anti-HNE antibodies. Furthermore, peptide sequencing and database searching revealed 95 distinct HNE-modified proteins and their exact modification sites, with 88 protein adducts being unique to OA chondrocytes. HNE-proteins of specific interest included histone H2A, H2B and H4, collagen alpha-3(VI) chain, eukaryotic initiation factor 4A-I, and nucleolar RNA helicase 2. Comparing their MS/MS spectra to those of HNE-modified standard peptides further validated the six HNE-proteins. SIGNIFICANCE: HNE binding to proteins has been shown to result in multiple abnormalities of chondrocyte phenotype and function, suggesting its contribution in OA development. Considering the increased levels of HNE in OA cartilage, this reactive aldehyde could play a role in OA. This work represents a clinically-relevant in vivo study to demonstrate the pathophysiological role of HNE in human OA. Since HNE binding can alter protein conformation and function, it remains highly relevant to study the effects of this modification in OA.
Collapse
Affiliation(s)
- Timon Geib
- Chemistry Department, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Cristiana Iacob
- Orthopaedic Research Laboratory, Hôpital du Sacré-Coeur, University of Montréal, Montréal, Québec, Canada
| | - Rihab Jribi
- Orthopaedic Research Laboratory, Hôpital du Sacré-Coeur, University of Montréal, Montréal, Québec, Canada
| | - Julio Fernandes
- Orthopaedic Research Laboratory, Hôpital du Sacré-Coeur, University of Montréal, Montréal, Québec, Canada
| | - Mohamed Benderdour
- Orthopaedic Research Laboratory, Hôpital du Sacré-Coeur, University of Montréal, Montréal, Québec, Canada.
| | - Lekha Sleno
- Chemistry Department, Université du Québec à Montréal, Montréal, Québec, Canada.
| |
Collapse
|
10
|
Bass AKA, El-Zoghbi MS, Nageeb ESM, Mohamed MFA, Badr M, Abuo-Rahma GEDA. Comprehensive review for anticancer hybridized multitargeting HDAC inhibitors. Eur J Med Chem 2020; 209:112904. [PMID: 33077264 DOI: 10.1016/j.ejmech.2020.112904] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/18/2020] [Accepted: 09/30/2020] [Indexed: 02/08/2023]
Abstract
Despite the encouraging clinical progress of chemotherapeutic agents in cancer treatment, innovation and development of new effective anticancer candidates still represents a challenging endeavor. With 15 million death every year in 2030 according to the estimates, cancer has increased rising of an alarm as a real crisis for public health and health systems worldwide. Therefore, scientist began to introduce innovative solutions to control the cancer global health problem. One of the promising strategies in this issue is the multitarget or smart hybrids having two or more pharmacophores targeting cancer. These rationalized hybrid molecules have gained great interests in cancer treatment as they are capable to simultaneously inhibit more than cancer pathway or target without drug-drug interactions and with less side effects. A prime important example of these hybrids, the HDAC hybrid inhibitors or referred as multitargeting HDAC inhibitors. The ability of HDAC inhibitors to synergistically improve the efficacy of other anti-cancer drugs and moreover, the ease of HDAC inhibitors cap group modification prompt many medicinal chemists to innovate and develop new generation of HDAC hybrid inhibitors. Notably, and during this short period, there are four HDAC inhibitor hybrids have entered different phases of clinical trials for treatment of different types of blood and solid tumors, namely; CUDC-101, CUDC-907, Tinostamustine, and Domatinostat. This review shed light on the most recent hybrids of HDACIs with one or more other cancer target pharmacophore. The designed multitarget hybrids include topoisomerase inhibitors, kinase inhibitors, nitric oxide releasers, antiandrogens, FLT3 and JAC-2 inhibitors, PDE5-inhibitors, NAMPT-inhibitors, Protease inhibitors, BRD4-inhibitors and other targets. This review may help researchers in development and discovery of new horizons in cancer treatment.
Collapse
Affiliation(s)
- Amr K A Bass
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Menoufia University, Menoufia, Egypt
| | - Mona S El-Zoghbi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Menoufia University, Menoufia, Egypt
| | - El-Shimaa M Nageeb
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Mamdouh F A Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, 82524 Sohag, Egypt
| | - Mohamed Badr
- Department of Biochemistry, Faculty of Pharmacy, Menoufia University, Menoufia, Egypt
| | - Gamal El-Din A Abuo-Rahma
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University, New Minia, Minia, Egypt.
| |
Collapse
|
11
|
Vaidya GN, Rana P, Venkatesh A, Chatterjee DR, Contractor D, Satpute DP, Nagpure M, Jain A, Kumar D. Paradigm shift of "classical" HDAC inhibitors to "hybrid" HDAC inhibitors in therapeutic interventions. Eur J Med Chem 2020; 209:112844. [PMID: 33143937 DOI: 10.1016/j.ejmech.2020.112844] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
'Epigenetic' regulation of genes via post-translational modulation of proteins is the current mainstay approach for the disease therapies, particularly explored in the Histone Deacetylase (HDAC) class of enzymes. Mainly sight saw in cancer chemotherapeutics, HDAC inhibitors have also found a promising role in other diseases (neurodegenerative disorders, cardiovascular diseases, and viral infections) and successfully entered in various combination therapies (pre-clinical/clinical stages). The prevalent flexibility in the structural design of HDAC inhibitors makes them easily tuneable to merge with other pharmacophore modules for generating multi-targeted single hybrids as a novel tactic to overcome drawbacks of polypharmacy. Herein, we reviewed the putative role of prevalent HDAC hybrids inhibitors in the current and prospective stage as a translational approach to overcome the limitations of the existing conventional drug candidates (parent molecule) when used either alone (drug resistance, solubility issues, adverse side effects, selectivity profile) or in combination (pharmacokinetic interactions, patient compliance) for treating various diseases.
Collapse
Affiliation(s)
- Gargi Nikhil Vaidya
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Pooja Rana
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Ashwini Venkatesh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Deep Rohan Chatterjee
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Darshan Contractor
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Dinesh Parshuram Satpute
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Mithilesh Nagpure
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Alok Jain
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India; Department of Bio-Engineering, Birla Institute of Technology, Mesra, Ranchi, India.
| | - Dinesh Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
12
|
Stem cells and new intervention measures as emerging therapy in cardiac surgery. POLISH JOURNAL OF THORACIC AND CARDIOVASCULAR SURGERY 2020; 17:1-7. [PMID: 32728355 PMCID: PMC7379210 DOI: 10.5114/kitp.2020.94183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/20/2020] [Indexed: 11/17/2022]
Abstract
Cardiovascular disease (CVD) presents a great burden for elderly patients, their caregivers, and health systems. Structural and functional alterations of vessels accumulate throughout life, culminating in increased risk of developing CVD. Several inflammatory pathway are involved in vascular ageing. The growing elderly population worldwide highlights the need to understand how aging promotes CVD in order to develop new strategies to confront this challenge. In this review we analyzed the role of stem cells and new intervention measures as emerging drugs for vascular aging.
Collapse
|
13
|
Sánchez OF, Rodríguez AV, Velasco-España JM, Murillo LC, Sutachan JJ, Albarracin SL. Role of Connexins 30, 36, and 43 in Brain Tumors, Neurodegenerative Diseases, and Neuroprotection. Cells 2020; 9:E846. [PMID: 32244528 PMCID: PMC7226843 DOI: 10.3390/cells9040846] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/15/2020] [Accepted: 02/24/2020] [Indexed: 02/07/2023] Open
Abstract
Gap junction (GJ) channels and their connexins (Cxs) are complex proteins that have essential functions in cell communication processes in the central nervous system (CNS). Neurons, astrocytes, oligodendrocytes, and microglial cells express an extraordinary repertory of Cxs that are important for cell to cell communication and diffusion of metabolites, ions, neurotransmitters, and gliotransmitters. GJs and Cxs not only contribute to the normal function of the CNS but also the pathological progress of several diseases, such as cancer and neurodegenerative diseases. Besides, they have important roles in mediating neuroprotection by internal or external molecules. However, regulation of Cx expression by epigenetic mechanisms has not been fully elucidated. In this review, we provide an overview of the known mechanisms that regulate the expression of the most abundant Cxs in the central nervous system, Cx30, Cx36, and Cx43, and their role in brain cancer, CNS disorders, and neuroprotection. Initially, we focus on describing the Cx gene structure and how this is regulated by epigenetic mechanisms. Then, the posttranslational modifications that mediate the activity and stability of Cxs are reviewed. Finally, the role of GJs and Cxs in glioblastoma, Alzheimer's, Parkinson's, and Huntington's diseases, and neuroprotection are analyzed with the aim of shedding light in the possibility of using Cx regulators as potential therapeutic molecules.
Collapse
Affiliation(s)
- Oscar F. Sánchez
- Department of Nutrition and Biochemistry, Pontificia Universidad Javeriana, 110911 Bogota, Colombia; (A.V.R.); (J.M.V.-E.); (L.C.M.); (J.-J.S.)
| | | | | | | | | | - Sonia-Luz Albarracin
- Department of Nutrition and Biochemistry, Pontificia Universidad Javeriana, 110911 Bogota, Colombia; (A.V.R.); (J.M.V.-E.); (L.C.M.); (J.-J.S.)
| |
Collapse
|
14
|
Guarner-Lans V, Ramírez-Higuera A, Rubio-Ruiz ME, Castrejón-Téllez V, Soto ME, Pérez-Torres I. Early Programming of Adult Systemic Essential Hypertension. Int J Mol Sci 2020; 21:E1203. [PMID: 32054074 PMCID: PMC7072742 DOI: 10.3390/ijms21041203] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/27/2020] [Accepted: 02/10/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases are being included in the study of developmental origins of health and disease (DOHaD) and essential systemic hypertension has also been added to this field. Epigenetic modifications are one of the main mechanisms leading to early programming of disease. Different environmental factors occurring during critical windows in the early stages of life may leave epigenetic cues, which may be involved in the programming of hypertension when individuals reach adulthood. Such environmental factors include pre-term birth, low weight at birth, altered programming of different organs such as the blood vessels and the kidney, and living in disadvantageous conditions in the programming of hypertension. Mechanisms behind these factors that impact on the programming include undernutrition, oxidative stress, inflammation, emotional stress, and changes in the microbiota. These factors and their underlying causes acting at the vascular level will be discussed in this paper. We also explore the establishment of epigenetic cues that may lead to hypertension at the vascular level such as DNA methylation, histone modifications (methylation and acetylation), and the role of microRNAs in the endothelial cells and blood vessel smooth muscle which participate in hypertension. Since epigenetic changes are reversible, the knowledge of this type of markers could be useful in the field of prevention, diagnosis or epigenetic drugs as a therapeutic approach to hypertension.
Collapse
Affiliation(s)
- Verónica Guarner-Lans
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City 14080, Mexico; (M.E.R.-R.); (V.C.-T.)
| | - Abril Ramírez-Higuera
- Nutrition Biochemistry Laboratory, Research and Food Development Unit. Veracruz Technological Institute, National Technological of Mexico, Veracruz 91897, Mexico;
| | - María Esther Rubio-Ruiz
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City 14080, Mexico; (M.E.R.-R.); (V.C.-T.)
| | - Vicente Castrejón-Téllez
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City 14080, Mexico; (M.E.R.-R.); (V.C.-T.)
| | - María Elena Soto
- Department of Immunology, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico 14080, Mexico;
| | - Israel Pérez-Torres
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico 14080, Mexico;
| |
Collapse
|
15
|
Sangwan R, Rajan R, Mandal PK. HDAC as onco target: Reviewing the synthetic approaches with SAR study of their inhibitors. Eur J Med Chem 2018; 158:620-706. [DOI: 10.1016/j.ejmech.2018.08.073] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/09/2018] [Accepted: 08/26/2018] [Indexed: 02/06/2023]
|
16
|
Nitric oxide and the biology of pregnancy. Vascul Pharmacol 2018; 110:71-74. [PMID: 30076925 DOI: 10.1016/j.vph.2018.07.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 07/29/2018] [Indexed: 01/16/2023]
Abstract
Nitric oxide (NO) is a key regulator of both maternal and fetal homeostasis during pregnancy, facilitating the maternal cardio-vascular changes, fetal development and growth and adaptation to extrauterine life. Dysfunction of the NO system during pregnancy is associated to placental and vascular-related diseases such as hypertensive disorders of pregnancy (HDP) and intrauterine growth restriction (IUGR). Emerging therapeutic strategies involving NO precursors, NO donors, natural derivatives or pharmacological modulators of the NO system seem hold promise for the treatment of such conditions of pregnancy.
Collapse
|
17
|
Sato A, Kim JD, Mizukami H, Nakashima M, Kako K, Ishida J, Itakura A, Takeda S, Fukamizu A. Gestational changes in PRMT1 expression of murine placentas. Placenta 2018; 65:47-54. [PMID: 29908641 DOI: 10.1016/j.placenta.2018.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 03/24/2018] [Accepted: 04/06/2018] [Indexed: 10/17/2022]
Abstract
INTRODUCTION In mammals, the placenta is an organ that is required to maintain the development of fetus during pregnancy. Although the proper formation of placenta is in part regulated by the post-translational modifications of proteins, little is known regarding protein arginine methylation during placental development. Here, we characterized developmental expression of protein arginine methyltransferase 1 (PRMT1) in mouse placentas. METHODS Expression levels of PRMT1 mRNA and protein in placentas were investigated using the real-time quantitative PCR and Western blot, respectively. Next, the localization of PRMT1 was determined by immunohistochemistry and immunofluorescence analyses. In addition, the levels of methylarginines of placental proteins were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS PRMT1 mRNA and its protein were expressed at highest levels in mid-gestation stages, and their expression showed stepwise decrease in the late gestation. At embryonic (E) day 9, PRMT1 was observed in several different trophoblast cell (TC) subtypes. Furthermore, PRMT1 was mainly expressed in the labyrinth zone of TCs at E13. Finally, total methylarginines of proteins were significantly reduced in late gestation of placentas compared with mid-gestation stages. DISCUSSION In this study, we found developmental changes in the placental expression of PRMT1 and in protein arginine methylation status during pregnancy. These findings provide fundamental information regarding placental PRMT1-mediated arginine methylation during the development.
Collapse
Affiliation(s)
- Anna Sato
- Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo 113-8421, Japan
| | - Jun-Dal Kim
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Hayase Mizukami
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| | - Misaki Nakashima
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| | - Koichiro Kako
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| | - Junji Ishida
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Atsuo Itakura
- Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo 113-8421, Japan
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo 113-8421, Japan
| | - Akiyoshi Fukamizu
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan.
| |
Collapse
|
18
|
Owusu Darkwa E, Djagbletey R, Sottie D, Owoo C, Vanderpuye NM, Essuman R, Aryee G. Serum nitric oxide levels in healthy pregnant women: a case- control study in a tertiary facility in Ghana. Matern Health Neonatol Perinatol 2018; 4:3. [PMID: 29479454 PMCID: PMC5819155 DOI: 10.1186/s40748-017-0072-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/28/2017] [Indexed: 12/31/2022] Open
Abstract
Background Pregnancy is associated with significant changes in maternal cardiovascular system which regulates oxygen and nutrient supply to the growing foetus. Nitric oxide, a physiologic vascular smooth muscle relaxant regulates blood flow and therefore may play a role in the cardiovascular changes in pregnancy. The study aimed to determine the levels and changes in maternal serum nitric oxide levels during healthy pregnancy. Methods A case-control study was conducted among 32 healthy non-pregnant women as controls and 100 healthy pregnant women (consisting of 33 first trimester, 37 s trimester, and 30 third trimester) as cases. Subjects were consecutively recruited into the study after obtaining an informed consent and meeting the inclusion criteria. Griess Reagent method was used to determine serum nitric oxide levels. Results There were no statistically significant difference in the ages and parity of recruited cases and controls. Mean arterial blood pressures were significantly lower (p = 0.009) and serum nitric oxide levels were significantly higher (p < 0.001) in healthy pregnant women compared to healthy non-pregnant women. There was a non-significant progressive increase in serum nitric oxide levels during healthy normal pregnancy. Conclusions The finding of a significantly reduced blood pressures and a significant increase in serum nitric oxide levels in healthy pregnancy may suggest a role of nitric oxide in vascular adaptation in pregnancy.
Collapse
Affiliation(s)
- Ebenezer Owusu Darkwa
- 1Department of Anaesthesia, Korle-bu Teaching Hospital, University of Ghana School of Medicine and Dentistry, College of Health Sciences, P. O. Box 4236, Accra, Ghana
| | - Robert Djagbletey
- 1Department of Anaesthesia, Korle-bu Teaching Hospital, University of Ghana School of Medicine and Dentistry, College of Health Sciences, P. O. Box 4236, Accra, Ghana
| | - Daniel Sottie
- 2Department of Anaesthesia, Korle-Bu Teaching Hospital, Accra, Ghana
| | - Christian Owoo
- 1Department of Anaesthesia, Korle-bu Teaching Hospital, University of Ghana School of Medicine and Dentistry, College of Health Sciences, P. O. Box 4236, Accra, Ghana
| | | | - Raymond Essuman
- 1Department of Anaesthesia, Korle-bu Teaching Hospital, University of Ghana School of Medicine and Dentistry, College of Health Sciences, P. O. Box 4236, Accra, Ghana
| | - George Aryee
- 1Department of Anaesthesia, Korle-bu Teaching Hospital, University of Ghana School of Medicine and Dentistry, College of Health Sciences, P. O. Box 4236, Accra, Ghana
| |
Collapse
|
19
|
Structural and biological characterization of new hybrid drugs joining an HDAC inhibitor to different NO-donors. Eur J Med Chem 2018; 144:612-625. [DOI: 10.1016/j.ejmech.2017.12.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/10/2017] [Accepted: 12/13/2017] [Indexed: 01/23/2023]
|
20
|
Pambianco S, Giovarelli M, Perrotta C, Zecchini S, Cervia D, Di Renzo I, Moscheni C, Ripolone M, Violano R, Moggio M, Bassi MT, Puri PL, Latella L, Clementi E, De Palma C. Reversal of Defective Mitochondrial Biogenesis in Limb-Girdle Muscular Dystrophy 2D by Independent Modulation of Histone and PGC-1α Acetylation. Cell Rep 2017; 17:3010-3023. [PMID: 27974213 DOI: 10.1016/j.celrep.2016.11.044] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 06/10/2016] [Accepted: 11/11/2016] [Indexed: 01/05/2023] Open
Abstract
Mitochondrial dysfunction occurs in many muscle degenerative disorders. Here, we demonstrate that mitochondrial biogenesis was impaired in limb-girdle muscular dystrophy (LGMD) 2D patients and mice and was associated with impaired OxPhos capacity. Two distinct approaches that modulated histones or peroxisome proliferator-activated receptor-gamma coactivator 1 α (PGC-1α) acetylation exerted equivalent functional effects by targeting different mitochondrial pathways (mitochondrial biogenesis or fatty acid oxidation[FAO]). The histone deacetylase inhibitor Trichostatin A (TSA) changed chromatin assembly at the PGC-1α promoter, restored mitochondrial biogenesis, and enhanced muscle oxidative capacity. Conversely, nitric oxide (NO) triggered post translation modifications of PGC-1α and induced FAO, recovering the bioenergetics impairment of muscles but shunting the defective mitochondrial biogenesis. In conclusion, a transcriptional blockade of mitochondrial biogenesis occurred in LGMD-2D and could be recovered by TSA changing chromatin conformation, or it could be overcome by NO activating a mitochondrial salvage pathway.
Collapse
Affiliation(s)
- Sarah Pambianco
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences, Unit of Clinical Pharmacology, University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco, National Research Council-Institute of Neuroscience, Università degli Studi di Milano, 20157 Milano, Italy
| | - Davide Cervia
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy; Department for Innovation in Biological, Agro-food and Forest systems, Università degli Studi della Tuscia, 01100 Viterbo, Italy
| | - Ilaria Di Renzo
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy
| | - Michela Ripolone
- Neuromuscular Unit, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, 20122 Milano, Italy
| | - Raffaella Violano
- Neuromuscular Unit, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, 20122 Milano, Italy
| | - Maurizio Moggio
- Neuromuscular Unit, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, 20122 Milano, Italy
| | | | - Pier Lorenzo Puri
- Epigenetics and Regenerative Pharmacology, IRCCS Fondazione Santa Lucia, 00142 Roma, Italy; Sanford Children's Health Research Center, Sanford Prebys Burnham Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Lucia Latella
- Epigenetics and Regenerative Pharmacology, IRCCS Fondazione Santa Lucia, 00142 Roma, Italy; National Research Council-Institute of Translational Pharmacology, 00179 Roma, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences, Unit of Clinical Pharmacology, University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco, National Research Council-Institute of Neuroscience, Università degli Studi di Milano, 20157 Milano, Italy; IRCCS Eugenio Medea, 23842 Bosisio Parini, Italy.
| | - Clara De Palma
- Department of Biomedical and Clinical Sciences, Unit of Clinical Pharmacology, University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco, National Research Council-Institute of Neuroscience, Università degli Studi di Milano, 20157 Milano, Italy.
| |
Collapse
|
21
|
Pineda E, Perdomo D. Entamoeba histolytica under Oxidative Stress: What Countermeasure Mechanisms Are in Place? Cells 2017; 6:cells6040044. [PMID: 29160807 PMCID: PMC5755502 DOI: 10.3390/cells6040044] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/17/2017] [Accepted: 11/17/2017] [Indexed: 02/06/2023] Open
Abstract
Entamoeba histolytica is the causative agent of human amoebiasis; it affects 50 million people worldwide and causes approximately 100,000 deaths per year. Entamoeba histolytica is an anaerobic parasite that is primarily found in the colon; however, for unknown reasons, it can become invasive, breaching the gut barrier and migrating toward the liver causing amoebic liver abscesses. During the invasive process, it must maintain intracellular hypoxia within the oxygenated human tissues and cellular homeostasis during the host immune defense attack when it is confronted with nitric oxide and reactive oxygen species. But how? This review will address the described and potential mechanisms available to counter the oxidative stress generated during invasion and the possible role that E. histolytica’s continuous endoplasmic reticulum (Eh-ER) plays during these events.
Collapse
Affiliation(s)
- Erika Pineda
- Laboratory of Fundamental Microbiology and Pathogenicity (MFP), University of Bordeaux, CNRS UMR-5234, 33000 Bordeaux, France.
| | - Doranda Perdomo
- Laboratory of Fundamental Microbiology and Pathogenicity (MFP), University of Bordeaux, CNRS UMR-5234, 33000 Bordeaux, France.
| |
Collapse
|
22
|
Deryagin OG, Gavrilova SA, Gainutdinov KL, Golubeva AV, Andrianov VV, Yafarova GG, Buravkov SV, Koshelev VB. Molecular Bases of Brain Preconditioning. Front Neurosci 2017; 11:427. [PMID: 28790886 PMCID: PMC5524930 DOI: 10.3389/fnins.2017.00427] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/11/2017] [Indexed: 12/20/2022] Open
Abstract
Preconditioning of the brain induces tolerance to the damaging effects of ischemia and prevents cell death in ischemic penumbra. The development of this phenomenon is mediated by mitochondrial adenosine triphosphate-sensitive potassium (KATP+) channels and nitric oxide signaling (NO). The aim of this study was to investigate the dynamics of molecular changes in mitochondria after ischemic preconditioning (IP) and the effect of pharmacological preconditioning (PhP) with the KATP+-channels opener diazoxide on NO levels after ischemic stroke in rats. Immunofluorescence-histochemistry and laser-confocal microscopy were applied to evaluate the cortical expression of electron transport chain enzymes, mitochondrial KATP+-channels, neuronal and inducible NO-synthases, as well as the dynamics of nitrosylation and nitration of proteins in rats during the early and delayed phases of IP. NO cerebral content was studied with electron paramagnetic resonance (EPR) spectroscopy using spin trapping. We found that 24 h after IP in rats, there is a two-fold decrease in expression of mitochondrial KATP+-channels (p = 0.012) in nervous tissue, a comparable increase in expression of cytochrome c oxidase (p = 0.008), and a decrease in intensity of protein S-nitrosylation and nitration (p = 0.0004 and p = 0.001, respectively). PhP led to a 56% reduction of free NO concentration 72 h after ischemic stroke simulation (p = 0.002). We attribute this result to the restructuring of tissue energy metabolism, namely the provision of increased catalytic sites to mitochondria and the increased elimination of NO, which prevents a decrease in cell sensitivity to oxygen during subsequent periods of severe ischemia.
Collapse
Affiliation(s)
- Oleg G Deryagin
- Department of Physiology and General Pathology, Medical Faculty, Lomonosov Moscow State UniversityMoscow, Russia
| | - Svetlana A Gavrilova
- Department of Physiology and General Pathology, Medical Faculty, Lomonosov Moscow State UniversityMoscow, Russia
| | - Khalil L Gainutdinov
- Laboratory of Neurorehabilitation of Motor Disorders, Institute of Fundamental Medicine and Biology, Kazan Federal UniversityKazan, Russia.,Laboratory of Spin Physics and Spin Chemistry, Zavoisky Physical-Technical Institute of the Russian Academy of SciencesKazan, Russia
| | - Anna V Golubeva
- Department of Physiology and General Pathology, Medical Faculty, Lomonosov Moscow State UniversityMoscow, Russia
| | - Vyatcheslav V Andrianov
- Laboratory of Neurorehabilitation of Motor Disorders, Institute of Fundamental Medicine and Biology, Kazan Federal UniversityKazan, Russia.,Laboratory of Spin Physics and Spin Chemistry, Zavoisky Physical-Technical Institute of the Russian Academy of SciencesKazan, Russia
| | - Guzel G Yafarova
- Laboratory of Neurorehabilitation of Motor Disorders, Institute of Fundamental Medicine and Biology, Kazan Federal UniversityKazan, Russia.,Laboratory of Spin Physics and Spin Chemistry, Zavoisky Physical-Technical Institute of the Russian Academy of SciencesKazan, Russia
| | - Sergey V Buravkov
- Research Laboratory of Cellular Structure and Tissue Imaging Analysis, Medical Faculty, Lomonosov Moscow State UniversityMoscow, Russia
| | - Vladimir B Koshelev
- Department of Physiology and General Pathology, Medical Faculty, Lomonosov Moscow State UniversityMoscow, Russia
| |
Collapse
|
23
|
Barbati SA, Colussi C, Bacci L, Aiello A, Re A, Stigliano E, Isidori AM, Grassi C, Pontecorvi A, Farsetti A, Gaetano C, Nanni S. Transcription Factor CREM Mediates High Glucose Response in Cardiomyocytes and in a Male Mouse Model of Prolonged Hyperglycemia. Endocrinology 2017; 158:2391-2405. [PMID: 28368536 DOI: 10.1210/en.2016-1960] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/16/2017] [Indexed: 01/31/2023]
Abstract
This study aims at investigating the epigenetic landscape of cardiomyocytes exposed to elevated glucose levels. High glucose (30 mM) for 72 hours determined some epigenetic changes in mouse HL-1 and rat differentiated H9C2 cardiomyocytes including upregulation of class I and III histone deacetylase protein levels and activity, inhibition of histone acetylase p300 activity, increase in histone H3 lysine 27 trimethylation, and reduction in H3 lysine 9 acetylation. Gene expression analysis focused on cardiotoxicity revealed that high glucose induced markers associated with tissue damage, fibrosis, and cardiac remodeling such as Nexilin (NEXN), versican, cyclic adenosine 5'-monophosphate-responsive element modulator (CREM), and adrenoceptor α2A (ADRA2). Notably, the transcription factor CREM was found to be important in the regulation of cardiotoxicity-associated genes as assessed by specific small interfering RNA and chromatin immunoprecipitation experiments. In CD1 mice, made hyperglycemic by streptozotoicin (STZ) injection, cardiac structural alterations were evident at 6 months after STZ treatment and were associated with a significant increase of H3 lysine 27 trimethylation and reduction of H3 lysine 9 acetylation. Consistently, NEXN, CREM, and ADRA2 expression was significantly induced at the RNA and protein levels. Confocal microscopy analysis of NEXN localization showed this protein irregularly distributed along the sarcomeres in the heart of hyperglycemic mice. This evidence suggested a structural alteration of cardiac Z-disk with potential consequences on contractility. In conclusion, high glucose may alter the epigenetic landscape of cardiac cells. Sildenafil, restoring guanosine 3', 5'-cyclic monophosphate levels, counteracted the increase of CREM and NEXN, providing a protective effect in the presence of hyperglycemia.
Collapse
Affiliation(s)
- Saviana A Barbati
- Institute of Human Physiology, Università Cattolica di Roma, 00168 Rome, Italy
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
| | - Claudia Colussi
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
- Institute of Cell Biology and Neurobiology, National Research Council, 00143 Rome, Italy
| | - Lorenza Bacci
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
| | - Aurora Aiello
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
- Institute of Cell Biology and Neurobiology, National Research Council, 00143 Rome, Italy
| | - Agnese Re
- Institute of Cell Biology and Neurobiology, National Research Council, 00143 Rome, Italy
| | - Egidio Stigliano
- Department of Histopathology, Università Cattolica di Roma, 00168 Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica di Roma, 00168 Rome, Italy
| | - Alfredo Pontecorvi
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
| | - Antonella Farsetti
- Institute of Cell Biology and Neurobiology, National Research Council, 00143 Rome, Italy
- Medicine Clinic III, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Carlo Gaetano
- Medicine Clinic III, Division of Cardiovascular Epigenetics, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Simona Nanni
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
| |
Collapse
|
24
|
Cencioni C, Atlante S, Savoia M, Martelli F, Farsetti A, Capogrossi MC, Zeiher AM, Gaetano C, Spallotta F. The double life of cardiac mesenchymal cells: Epimetabolic sensors and therapeutic assets for heart regeneration. Pharmacol Ther 2016; 171:43-55. [PMID: 27742569 DOI: 10.1016/j.pharmthera.2016.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Organ-specific mesenchymal cells naturally reside in the stroma, where they are exposed to some environmental variables affecting their biology and functions. Risk factors such as diabetes or aging influence their adaptive response. In these cases, permanent epigenetic modifications may be introduced in the cells with important consequences on their local homeostatic activity and therapeutic potential. Numerous results suggest that mesenchymal cells, virtually present in every organ, may contribute to tissue regeneration mostly by paracrine mechanisms. Intriguingly, the heart is emerging as a source of different cells, including pericytes, cardiac progenitors, and cardiac fibroblasts. According to phenotypic, functional, and molecular criteria, these should be classified as mesenchymal cells. Not surprisingly, in recent years, the attention on these cells as therapeutic tools has grown exponentially, although only very preliminary data have been obtained in clinical trials to date. In this review, we summarized the state of the art about the phenotypic features, functions, regenerative properties, and clinical applicability of mesenchymal cells, with a particular focus on those of cardiac origin.
Collapse
Affiliation(s)
- Chiara Cencioni
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Sandra Atlante
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Matteo Savoia
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Universitá Cattolica, Institute of Medical Pathology, 00138 Rome, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, San Donato Milanese, Milan 20097, Italy.
| | - Antonella Farsetti
- Consiglio Nazionale delle Ricerche, Istituto di Biologia Cellulare e Neurobiologia, Roma, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Maurizio C Capogrossi
- Laboratorio di Patologia Vascolare, Istituto Dermopatico dell'Immacolata, Roma, Italy.
| | - Andreas M Zeiher
- Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Carlo Gaetano
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Francesco Spallotta
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| |
Collapse
|
25
|
Vasudevan D, Bovee RC, Thomas DD. Nitric oxide, the new architect of epigenetic landscapes. Nitric Oxide 2016; 59:54-62. [PMID: 27553128 DOI: 10.1016/j.niox.2016.08.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 08/18/2016] [Indexed: 12/13/2022]
Abstract
Nitric oxide (NO) is an endogenously produced signaling molecule with multiple regulatory functions in physiology and disease. The most studied molecular mechanisms underlying the biological functions of NO include its reaction with heme proteins and regulation of protein activity via modification of thiol residues. A significant number of transcriptional responses and phenotypes observed in NO microenvironments, however, still lack mechanistic understanding. Recent studies shed new light on NO signaling by revealing its influence on epigenetic changes within the cell. Epigenetic alterations are important determinants of transcriptional responses and cell phenotypes, which can relay heritable information during cell division. As transcription across the genome is highly sensitive to these upstream epigenetic changes, this mode of NO signaling provides an alternate explanation for NO-mediated gene expression changes and phenotypes. This review will provide an overview of the interplay between NO and epigenetics as well as emphasize the unprecedented importance of these pathways to explain phenotypic effects associated with biological NO synthesis.
Collapse
Affiliation(s)
- Divya Vasudevan
- Department of Urology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Rhea C Bovee
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Douglas D Thomas
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
26
|
Gasotransmitters in Gametogenesis and Early Development: Holy Trinity for Assisted Reproductive Technology-A Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1730750. [PMID: 27579148 PMCID: PMC4992752 DOI: 10.1155/2016/1730750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/03/2016] [Indexed: 11/23/2022]
Abstract
Creation of both gametes, sperm and oocyte, and their fusion during fertilization are essential step for beginning of life. Although molecular mechanisms regulating gametogenesis, fertilization, and early embryonic development are still subjected to intensive study, a lot of phenomena remain unclear. Based on our best knowledge and own results, we consider gasotransmitters to be essential for various signalisation in oocytes and embryos. In accordance with nitric oxide (NO) and hydrogen sulfide (H2S) physiological necessity, their involvement during oocyte maturation and regulative role in fertilization followed by embryonic development have been described. During these processes, NO- and H2S-derived posttranslational modifications represent the main mode of their regulative effect. While NO represent the most understood gasotransmitter and H2S is still intensively studied gasotransmitter, appreciation of carbon monoxide (CO) role in reproduction is still missing. Overall understanding of gasotransmitters including their interaction is promising for reproductive medicine and assisted reproductive technologies (ART), because these approaches contend with failure of in vitro assisted reproduction.
Collapse
|
27
|
Roche J, Bertrand P. Inside HDACs with more selective HDAC inhibitors. Eur J Med Chem 2016; 121:451-483. [PMID: 27318122 DOI: 10.1016/j.ejmech.2016.05.047] [Citation(s) in RCA: 245] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/20/2016] [Accepted: 05/21/2016] [Indexed: 01/08/2023]
Abstract
Inhibitors of histone deacetylases (HDACs) are nowadays part of the therapeutic arsenal mainly against cancers, with four compounds approved by the Food and Drug Administration. During the last five years, several groups have made continuous efforts to improve this class of compounds, designing more selective compounds or compounds with multiple capacities. After a survey of the HDAC biology and structures, this review summarizes the results of the chemists working in this field, and highlights when possible the behavior of the molecules inside their targets.
Collapse
Affiliation(s)
- Joëlle Roche
- Laboratoire Ecologie et Biologie des Interactions, Equipe « SEVE Sucres & Echanges Végétaux-Environnement », Université de Poitiers, UMR CNRS 7267, F-86073 Poitiers Cedex 09, France; Réseau Epigénétique du Cancéropôle Grand Ouest, France
| | - Philippe Bertrand
- Institut de Chimie des Milieux et Matériaux de Poitiers, UMR CNRS 7285, 4 rue Michel Brunet, TSA 51106, B28, F-86073 Poitiers Cedex 09, France; Réseau Epigénétique du Cancéropôle Grand Ouest, France.
| |
Collapse
|
28
|
Krause BJ, Hernandez C, Caniuguir A, Vasquez-Devaud P, Carrasco-Wong I, Uauy R, Casanello P. Arginase-2 is cooperatively up-regulated by nitric oxide and histone deacetylase inhibition in human umbilical artery endothelial cells. Biochem Pharmacol 2015; 99:53-9. [PMID: 26551598 DOI: 10.1016/j.bcp.2015.10.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/28/2015] [Indexed: 11/30/2022]
Abstract
Arginase-2 counteracts endothelial nitric oxide synthase (eNOS) activity in human endothelium, and its expression is negatively controlled by histone deacetylase (HDAC2). Conversely NO inhibits HDAC and previous studies suggest that arginase-2 is up-regulated by NO. We studied whether NO regulates arginase-2 expression in umbilical artery endothelial cells (HUAEC) increasing ARG2 promoter accessibility. HUAEC exposed to NOC-18 (NO donor, 1-100 μM, 0-24 h) showed an increase in arginase-2 but a decrease in eNOS mRNA levels in a time-dependent manner, with a maximal effect at 100 μM (24 h). Conversely NOS inhibition with L-NAME (100 μM) reduced arginase-2 mRNA and protein levels, an effect reverted by co-incubation with NOC-18. Treatment with TSA paralleled the effects of NO on arginase-2 and eNOS at mRNA and protein levels, with maximal effect at 10 μM. Co-incubation of NOC-18 (100 μM) with a sub-maximal concentration of TSA (1 μM) potentiated the increase in arginase-2 mRNA levels, whilst L-NAME prevented TSA-dependent arginase-2 induction. The effects on arginase-2 mRNA were paralleled by changes in chromatin accessibility, as well as increased levels of H3K9 and H4K12 acetylation, at ARG2 proximal (-579 to -367 and -280 to -73 bp from TSS) and core (-121 to +126 bp from TSS) promoter. Finally NO-dependent arginase-2 induction was prevented by pre-incubation for 10 min with the cysteine blocker MMTS (10 mM). These data showed for the first time that NO up-regulates arginase-2 expression in primary cultured human endothelial cells by an epigenetic-mediated mechanism increasing ARG2 promoter accessibility suggesting a negative regulatory loop for eNOS activity.
Collapse
Affiliation(s)
- Bernardo J Krause
- Division of Obstetrics & Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Cherie Hernandez
- Division of Obstetrics & Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andres Caniuguir
- Division of Obstetrics & Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paola Vasquez-Devaud
- Division of Obstetrics & Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ivo Carrasco-Wong
- Division of Obstetrics & Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ricardo Uauy
- Division of Pediatrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paola Casanello
- Division of Obstetrics & Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile; Division of Pediatrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
29
|
Duan W, Hou J, Chu X, Li X, Zhang J, Li J, Xu W, Zhang Y. Synthesis and biological evaluation of novel histone deacetylases inhibitors with nitric oxide releasing activity. Bioorg Med Chem 2015; 23:4481-4488. [PMID: 26122774 DOI: 10.1016/j.bmc.2015.06.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/04/2015] [Accepted: 06/05/2015] [Indexed: 11/21/2022]
Abstract
A novel series of histone deacetylases inhibitors (HDACIs) containing benzofuroxan pharmacophore as nitric oxide (NO) donor were designed based on the combination principle and 'multifunctional drugs' theory. As a novel study on embedding NO donor into the structure of HDACIs, all designed hybrid compounds, especially 19d and 24d, displayed remarkable HDACs inhibitory activity and outstanding antiproliferative activity on tumor cells. Besides, they could produce high levels of NO in HCT-116 cells; furthermore, their antiproliferative activity on HCT-116 cells could be diminished by pretreatment with hemoglobin, as the NO scavenger, in a dose-dependent manner. All in all, our designed compounds displayed great inhibitory activities and might offer a prospective avenue to discover novel anti-cancer drugs.
Collapse
Affiliation(s)
- Wenwen Duan
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Ji'nan, Shandong 250012, People's Republic of China
| | - Jinning Hou
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Ji'nan, Shandong 250012, People's Republic of China
| | - Xiaojing Chu
- Weifang Bochuang International Biological Medicinal Institute, Weifang, Shandong 261061, People's Republic of China
| | - Xiaoqian Li
- Color Ultrasonic Room, The People's Hospital of Shouguang, Weifang, Shandong 262700, People's Republic of China
| | - Jian Zhang
- Department of Medical Chemistry, School of Pharmacy, Weifang Medical University, West Baotong Street, Weifang, Shandong 261053, People's Republic of China
| | - Jin Li
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Ji'nan, Shandong 250012, People's Republic of China
| | - Wenfang Xu
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Ji'nan, Shandong 250012, People's Republic of China
| | - Yingjie Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Ji'nan, Shandong 250012, People's Republic of China.
| |
Collapse
|
30
|
Puyaubert J, Baudouin E. New clues for a cold case: nitric oxide response to low temperature. PLANT, CELL & ENVIRONMENT 2014; 37:2623-30. [PMID: 24720833 DOI: 10.1111/pce.12329] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 05/18/2023]
Abstract
Low temperature is among the most frequent stresses met by plants during their lifespan, and a plant's ability to cold-acclimate is a determinant for further growth and development. Although intensive research has provided a good picture of the molecular and metabolic changes triggered by cold, the underlying regulatory mechanisms remain elusive and are thus being actively sought. Recent studies have shed light on the importance of nitric oxide (NO), a ubiquitous signalling molecule in eukaryotes, for plant tolerance to chilling and freezing. Indeed, NO formation following cold exposure has been reported in a range of plant species, and a series of proteins targeted by NO-based post-translational modifications have been identified. Moreover, key cold-regulated genes have been characterized as NO-dependent, suggesting the crucial importance of NO signalling for cold-responsive gene expression. This review provides a picture of our current understanding of the function of NO in the context of plant response to cold. Particular attention is dedicated to the open questions left by the fragmented data currently available concerning NO formation, transduction and biological significance for plant adaptation to low temperature.
Collapse
Affiliation(s)
- Juliette Puyaubert
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Biologie du Développement, F-75005, Paris, France; CNRS, UMR 7622, Biologie du Développement, F-75005, Paris, France
| | | |
Collapse
|
31
|
Jones LH, Narayanan A, Hett EC. Understanding and applying tyrosine biochemical diversity. MOLECULAR BIOSYSTEMS 2014; 10:952-69. [PMID: 24623162 DOI: 10.1039/c4mb00018h] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review highlights some of the recent advances made in our understanding of the diversity of tyrosine biochemistry and shows how this has inspired novel applications in numerous areas of molecular design and synthesis, including chemical biology and bioconjugation. The pathophysiological implications of tyrosine biochemistry will be presented from a molecular perspective and the opportunities for therapeutic intervention explored.
Collapse
Affiliation(s)
- Lyn H Jones
- Pfizer R&D, Chemical Biology Group, BioTherapeutics Chemistry, WorldWide Medicinal Chemistry, 200 Cambridge Park Drive, Cambridge, MA 02140, USA.
| | | | | |
Collapse
|
32
|
Abstract
Epigenetics refers to long-term modifications of gene activity that can be inherited, either somatically or transgenerationally, but that are independent of alterations in the primary base sequence of the organism's DNA. These changes can include chemical modifications of both the DNA bases and the proteins that associate with the DNA helices to form chromatin, the nucleic acid:protein complex of which the chromosomes are comprised. Epigenetic modifications can affect the accessibility of the DNA for transcription factors (the DNA-binding proteins that specify which genes are to be active or silent by modulating the recruitment of the transcriptional machinery that reads the information encoded in the sequence) and thereby regulate the expression of genes and alter the phenotype of the organism. Epigenetic marks can also be re-established following mitosis, allowing patterns of differential gene expression to be transmitted from one cell generation to the next, and can even be maintained through meiosis, allowing transgenerational transfer of regulatory cues. Unlike the information encoded in the DNA sequence, which is invariant between most cell types and over time, epigenetic information is tissue specific and can change in response to exogenous and endogenous perturbations. This responsive capacity enables a sensitive and reactive system that can optimize gene expression in relevant tissue in response to environmental change. The realization that organisms are capable of genetically 'reprograming' themselves as well as 'preprograming' future cells, and even future offspring to optimize gene expression for a given environment may have tremendous ramifications on our understanding of both acclimatization and adaptation to hypoxia.
Collapse
Affiliation(s)
- Carolyn J Brown
- 1 Department of Medical Genetics, Molecular Epigenetics Group, University of British Columbia , Vancouver, British Columbia, Canada
| | | |
Collapse
|
33
|
Hertz R, Ben Lulu S, Shahi P, Trebicz-Geffen M, Benhar M, Ankri S. Proteomic identification of S-nitrosylated proteins in the parasite Entamoeba histolytica by resin-assisted capture: insights into the regulation of the Gal/GalNAc lectin by nitric oxide. PLoS One 2014; 9:e91518. [PMID: 24626316 PMCID: PMC3953491 DOI: 10.1371/journal.pone.0091518] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/12/2014] [Indexed: 11/19/2022] Open
Abstract
Entamoeba histolytica is a gastrointestinal protozoan parasite that causes amebiasis, a disease which has a worldwide distribution with substantial morbidity and mortality. Nitrosative stress, which is generated by innate immune cells, is one of the various environmental challenges that E. histolytica encounters during its life cycle. Although the effects of nitric oxide (NO) on the regulation of gene expression in this parasite have been previously investigated, our knowledge on S-nitrosylated proteins in E.histolytica is lacking. In order to fill this knowledge gap, we performed a large-scale detection of S-nitrosylated (SNO) proteins in E.histolytica trophozoites that were treated with the NO donor, S-nitrosocysteine by resin-assisted capture (RAC). We found that proteins involved in glycolysis, gluconeogenesis, translation, protein transport, and adherence to target cells such as the heavy subunit of Gal/GalNac lectin are among the S-nitrosylated proteins that were enriched by SNO-RAC. We also found that the S-nitrosylated cysteine residues in the carbohydrate recognition domain (CRD) of Gal/GalNAc lectin impairs its function and contributes to the inhibition of E.histolytica adherence to host cells. Collectively, these results advance our understanding of the mechanism of reduced E.histolytica adherence to mammalian cells by NO and emphasize the importance of NO as a regulator of key physiological functions in E.histolytica.
Collapse
Affiliation(s)
- Rivka Hertz
- Department of Molecular Microbiology, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Shani Ben Lulu
- Department of Biochemistry, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Preeti Shahi
- Department of Molecular Microbiology, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Meirav Trebicz-Geffen
- Department of Molecular Microbiology, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Moran Benhar
- Department of Biochemistry, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Serge Ankri
- Department of Molecular Microbiology, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
- * E-mail:
| |
Collapse
|
34
|
Oronsky B, Fanger GR, Oronsky N, Knox S, Scicinski J. The implications of hyponitroxia in cancer. Transl Oncol 2014; 7:167-73. [PMID: 24731473 PMCID: PMC4101386 DOI: 10.1016/j.tranon.2014.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/07/2014] [Accepted: 01/30/2014] [Indexed: 01/31/2023] Open
Abstract
Tumors are spatially heterogeneous, with regions of relative hypoxia and normoxia. The tumor microenvironment is an important determinant of both tumor growth and response to a variety of cytotoxic and targeted therapies. In the tumor microenvironment, reactive oxygen species and nitric oxide (NO) are important mediators of the level of expression of many transcription factors and signaling cascades that affect tumor growth and responses to therapy. The primary objective of this review is to explore and discuss the seemingly dichotomous actions of NO in cancer biology as both a tumor promoter and suppressor with an emphasis on understanding the role of persistently low NO concentrations or hyponitroxia as a key mediator in tumor progression. This review will also discuss the potential role of hyponitroxia as a novel therapeutic target to treat cancer and outline an approach that provides new opportunities for pharmacological intervention.
Collapse
Affiliation(s)
| | | | | | - Susan Knox
- Department of Radiation Oncology, Stanford University Medical Center, Stanford, CA, USA
| | | |
Collapse
|
35
|
The Entamoeba histolytica Dnmt2 homolog (Ehmeth) confers resistance to nitrosative stress. EUKARYOTIC CELL 2014; 13:494-503. [PMID: 24562908 DOI: 10.1128/ec.00031-14] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Nitric oxide (NO) has antimicrobial properties against many pathogens due to its reactivity as an S-nitrosylating agent. It inhibits many of the key enzymes that are involved in the metabolism and virulence of the parasite Entamoeba histolytica through S-nitrosylation of essential cysteine residues. Very little information is available on the mechanism of resistance to NO by pathogens in general and by this parasite in particular. Here, we report that exposure of the parasites to S-nitrosoglutathione (GSNO), an NO donor molecule, strongly reduces their viability and protein synthesis. However, the deleterious effects of NO were significantly reduced in trophozoites overexpressing Ehmeth, the cytosine-5 methyltransferase of the Dnmt2 family. Since these trophozoites also exhibited high levels of tRNA(Asp) methylation, the high levels suggested that Ehmeth-mediated tRNA(Asp) methylation is part of the resistance mechanism to NO. We previously reported that enolase, another glycolytic enzyme, binds to Ehmeth and inhibits its activity. We observed that the amount of Ehmeth-enolase complex was significantly reduced in GSNO-treated E. histolytica, which explains the aforementioned increase of tRNA methylation. Specifically, we demonstrated via site-directed mutagenesis that cysteine residues 228 and 229 of Ehmeth are susceptible to S-nitrosylation and are crucial for Ehmeth binding to enolase and for Ehmeth-mediated resistance to NO. These results indicate that Ehmeth has a central role in the response of the parasite to NO, and they contribute to the growing evidence that NO is a regulator of epigenetic mechanisms.
Collapse
|
36
|
Tu S, Yuan H, Hu J, Zhao C, Chai R, Cao H. Design, Synthesis and Biological Evaluation of Nitro Oxide Donating N-Hydroxycinnamamide Derivatives as Histone Deacetylase Inhibitors. Chem Pharm Bull (Tokyo) 2014; 62:1185-91. [DOI: 10.1248/cpb.c14-00449] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shiliang Tu
- Department of Coloproctology, Zhejiang Provincial People's Hospital
| | - Hang Yuan
- Department of Coloproctology, Zhejiang Provincial People's Hospital
| | - Jie Hu
- Department of Pharmacy, Wenzhou Medical University
| | | | - Rui Chai
- Department of Coloproctology, Zhejiang Provincial People's Hospital
| | - Hongfeng Cao
- Department of Coloproctology, Zhejiang Provincial People's Hospital
| |
Collapse
|
37
|
Borretto E, Lazzarato L, Spallotta F, Cencioni C, D’Alessandra Y, Gaetano C, Fruttero R, Gasco A. Synthesis and Biological Evaluation of the First Example of NO-Donor Histone Deacetylase Inhibitor. ACS Med Chem Lett 2013; 4:994-9. [PMID: 24900596 DOI: 10.1021/ml400289e] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 09/04/2013] [Indexed: 11/30/2022] Open
Abstract
The NO-donor histone deacetylase inhibitor 2, formally obtained by joining Entinostat 1, a moderately selective Class I histone deacetylases (HDACs) inhibitor, to a 4-(methylaminomethyl)furoxan-3-carbonitrile scaffold, is described and its preliminary biological profile discussed. This hybrid regulates Classes I and II HDACs. Nitric oxide (NO) released by the compound activates soluble guanylate cyclase (sGC), causing Class II nuclear shuttling and chromatin modifications, with consequences on gene expression. The hybrid affects a number of micro-RNAs not modulated by its individual components; it promotes myogenic differentiation, inducing the formation of larger myotubes with significantly more nuclei per fiber, in a more efficient manner than the 1:1 mixture of its two components. The hybrid is an example of a new class of NO-donor HDACs now being developed, which should be of interest for treating a number of diseases.
Collapse
Affiliation(s)
- Emily Borretto
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Torino, Italy
| | - Loretta Lazzarato
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Torino, Italy
| | - Francesco Spallotta
- Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino—IRCCS, Via Carlo Parea 4, 20138 Milan, Italy
| | - Chiara Cencioni
- Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino—IRCCS, Via Carlo Parea 4, 20138 Milan, Italy
| | - Yuri D’Alessandra
- Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino—IRCCS, Via Carlo Parea 4, 20138 Milan, Italy
| | - Carlo Gaetano
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Roberta Fruttero
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Torino, Italy
| | - Alberto Gasco
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Torino, Italy
| |
Collapse
|
38
|
Oxidative stress and epigenetic regulation in ageing and age-related diseases. Int J Mol Sci 2013; 14:17643-63. [PMID: 23989608 PMCID: PMC3794746 DOI: 10.3390/ijms140917643] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 08/19/2013] [Accepted: 08/21/2013] [Indexed: 01/04/2023] Open
Abstract
Recent statistics indicate that the human population is ageing rapidly. Healthy, but also diseased, elderly people are increasing. This trend is particularly evident in Western countries, where healthier living conditions and better cures are available. To understand the process leading to age-associated alterations is, therefore, of the highest relevance for the development of new treatments for age-associated diseases, such as cancer, diabetes, Alzheimer and cardiovascular accidents. Mechanistically, it is well accepted that the accumulation of intracellular damage determined by reactive oxygen species (ROS) might orchestrate the progressive loss of control over biological homeostasis and the functional impairment typical of aged tissues. Here, we review how epigenetics takes part in the control of stress stimuli and the mechanisms of ageing physiology and physiopathology. Alteration of epigenetic enzyme activity, histone modifications and DNA-methylation is, in fact, typically associated with the ageing process. Specifically, ageing presents peculiar epigenetic markers that, taken altogether, form the still ill-defined “ageing epigenome”. The comprehension of mechanisms and pathways leading to epigenetic modifications associated with ageing may help the development of anti-ageing therapies.
Collapse
|
39
|
Kalhan SC, Wilson-Costello D. Prematurity and programming: contribution of neonatal Intensive Care Unit interventions. J Dev Orig Health Dis 2013; 4:121-33. [PMID: 25054678 PMCID: PMC4115292 DOI: 10.1017/s204017441200061x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Contemporary clinical practice for the care of the prematurely born babies has markedly improved their rates of survival so that most of these babies are expected to grow up to live a healthy functional life. Since the clinical follow-up is of short duration (years), only limited data are available to relate non-communicable diseases in adult life to events and interventions in the neonatal period. The major events that could have a programming effect include: (1) intrauterine growth restriction; (2) interruption of pregnancy with change in redox and reactive oxygen species (ROS) injury; (3) nutritional and pharmacological protocols for clinical care; and (4) nutritional care in the first 2 years resulting in accelerated weight gain. The available data are discussed in the context of perturbations in one carbon (methyl transfer) metabolism and its possible programming effects. Although direct evidence for genomic methylation is not available, clinical and experimental data on impact of redox and ROS, of low protein intake, excess methionine load and vitamin A, on methyl transfers are reviewed. The consequences of antenatal and postnatal administration of glucocorticoids are presented. Analysis of the correlates of insulin sensitivity at older age, suggests that premature birth is the major contributor, and is compounded by gain in weight during infancy. We speculate that premature interruption of pregnancy and neonatal interventions by affecting one carbon metabolism may cause programming effects on the immature baby. These can be additive to the effects of intrauterine environment (growth restriction) and are compounded by accelerated growth in early infancy.
Collapse
Affiliation(s)
- S C Kalhan
- 1 Department of Molecular Medicine, Cleveland Clinic, Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - D Wilson-Costello
- 3 Department of Neonatology, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
40
|
Spallotta F, Cencioni C, Straino S, Nanni S, Rosati J, Artuso S, Manni I, Colussi C, Piaggio G, Martelli F, Valente S, Mai A, Capogrossi MC, Farsetti A, Gaetano C. A nitric oxide-dependent cross-talk between class I and III histone deacetylases accelerates skin repair. J Biol Chem 2013; 288:11004-12. [PMID: 23463510 DOI: 10.1074/jbc.m112.441816] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In a mouse model of skin repair we found that the class I-IIa histone deacetylase inhibitor trichostatin A accelerated tissue regeneration. Unexpectedly, this effect was suppressed by Sirtinol, a class III histone deacetylase (HDAC) (sirtuin)-selective inhibitor. The role of sirtuins (SIRTs) was then investigated by using resveratrol and a novel SIRT1-2-3 activator, the MC2562 compound we synthesized recently. Both resveratrol and MC2562 were effective in accelerating wound repair. The local administration of natural or synthetic SIRT activators, in fact, significantly accelerated skin regeneration by increasing keratinocyte proliferation. In vitro experiments revealed that the activation of SIRTs stimulated keratinocyte proliferation via endothelial NO synthase phosphorylation and NO production. In this condition, the class I member HDAC2 was found S-nitrosylated on cysteine, a post-transduction modification associated with loss of activity and DNA binding capacity. After deacetylase inhibitor or SIRT activator treatment, ChIP showed, in fact, a significant HDAC2 detachment from the promoter region of insulin growth factor I (IGF-I), fibroblast growth factor 10 (FGF-10), and Epithelial Growth Factor (EGF), which may be the final recipients and effectors of the SIRT-NO-HDAC signaling cascade. Consistently, the effect of SIRT activators was reduced in the presence of NG-nitro-L-arginine methyl ester (L-NAME), a general inhibitor of NO synthesis. In conclusion, the NO-dependent cross-talk among class III and I histone deacetylases suggests an unprecedented signaling pathway important for skin repair.
Collapse
Affiliation(s)
- Francesco Spallotta
- Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino, Istituto di Ricerca e Cura a Carattere Scientifico, 20138 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Burke AJ, Sullivan FJ, Giles FJ, Glynn SA. The yin and yang of nitric oxide in cancer progression. Carcinogenesis 2013; 34:503-12. [PMID: 23354310 DOI: 10.1093/carcin/bgt034] [Citation(s) in RCA: 276] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Nitric oxide (NO) is a short-lived, pleiotropic molecule that affects numerous critical functions in the body. Presently, there are markedly conflicting findings in the literature regarding NO and its role in carcinogenesis and tumor progression. NO has been shown to have dichotomous effects on cellular proliferation, apoptosis, migration, invasion, angiogenesis and many other important processes in cancer biology. It has been shown to be both pro- and antitumorigenic, depending on the concentration and the tumor microenvironment in question. NO is generated by three isoforms of NO synthase (NOS) that are widely expressed and sometimes upregulated in human tumors. Due to its vast array of physiological functions, it presents a huge challenge to researchers to discover its true potential in cancer biology and consequently, its use in anticancer therapies. In this study, we review the current knowledge in this area, with an emphasis placed on NO modulation as an anticancer therapy, focusing on NO-donating drugs and NOS inhibitors.
Collapse
Affiliation(s)
- Amy J Burke
- Prostate Cancer Institute, National University of Ireland Galway, Dublin, Ireland.
| | | | | | | |
Collapse
|
42
|
Gonzalez D, Rojas A, Herrera MB, Conlan RS. iNOS activation regulates β-catenin association with its partners in endothelial cells. PLoS One 2012; 7:e52964. [PMID: 23285236 PMCID: PMC3532412 DOI: 10.1371/journal.pone.0052964] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 11/26/2012] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Signals that disrupt β-catenin association to cadherins may influence the translocation of β-catenin to the nucleus to regulate transcription. Post-translational modification of proteins is a signalling event that may lead to changes in structural conformation, association or function of the target proteins. NO and its derivatives induce nitration of proteins during inflammation. It has been described that animals treated with NO donors showed increased permeability due to modulation of VE-cadherin/catenin complex. We, therefore, aim to evaluate the effect of iNOS activation on the expression, nuclear localisation and function of β-catenin in endothelial cells. METHODOLOGY/PRINCIPAL FINDINGS Expression, nuclear localisation, post-translational modifications and function of β-catenin was analysed by cell fractionation, immunoprecipitation, immunoblots, QRT-PCR and permeability assays in murine endothelial cells (H5V). Influence of macrophage activation on expression of VE-cadherin/p120-catenin/β-catenin complex in co-cultured H5V cells was also assessed. Activation of macrophages to produce NO provoked a decrease in VE-cadherin/p120-catenin/β-catenin expression in H5V cells. Phosphorylation of β-catenin, p120-catenin and VE-cadherin, and reduction in the barrier properties of the cell monolayer was associated with iNOS induction. Moreover, high NO levels provoked nitration of β-catenin, and induced its translocation to the nucleus. In the nucleus of NOS activated cells, nitration levels of β-catenin influenced its association with TCF4 and p65 proteins. High levels of NO altered β-catenin mediated gene expression of NFκB and Wnt target genes without affecting cell viability. CONCLUSIONS NOS activity modulates β-catenin post-translational modifications, function and its association with different partners to promote endothelial cell survival. Therapeutic manipulation of iNOS levels may remove a critical cytoprotective mechanism of importance in tumour angiogenesis.
Collapse
Affiliation(s)
- Deyarina Gonzalez
- Centre for NanoHealth, College of Medicine, Swansea University, Swansea, United Kingdom.
| | | | | | | |
Collapse
|
43
|
Huang LT, Hsieh CS, Chang KA, Tain YL. Roles of nitric oxide and asymmetric dimethylarginine in pregnancy and fetal programming. Int J Mol Sci 2012. [PMID: 23203083 PMCID: PMC3509599 DOI: 10.3390/ijms131114606] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nitric oxide (NO) regulates placental blood flow and actively participates in trophoblast invasion and placental development. Asymmetric dimethylarginine (ADMA) can inhibit NO synthase, which generates NO. ADMA has been associated with uterine artery flow disturbances such as preeclampsia. Substantial experimental evidence has reliably supported the hypothesis that an adverse in utero environment plays a role in postnatal physiological and pathophysiological programming. Growing evidence suggests that the placental nitrergic system is involved in epigenetic fetal programming. In this review, we discuss the roles of NO and ADMA in normal and compromised pregnancies as well as the link between placental insufficiency and epigenetic fetal programming.
Collapse
Affiliation(s)
- Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan; E-Mail:
- Department of Traditional Chinese Medicine, Chang Gung University, Linkow 333, Taiwan
| | - Chih-Sung Hsieh
- Department of Pediatric Surgery, Pingtung Christian Hospital, Pingtung 900, Taiwan; E-Mail:
- Department of Nursing, MeiHo University, Pingtung 900, Taiwan
| | - Kow-Aung Chang
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan; E-Mail:
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan; E-Mail:
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-975056995; Fax: +886-7-7338009
| |
Collapse
|
44
|
Colussi C, Scopece A, Vitale S, Spallotta F, Mattiussi S, Rosati J, Illi B, Mai A, Castellano S, Sbardella G, Farsetti A, Capogrossi MC, Gaetano C. P300/CBP associated factor regulates nitroglycerin-dependent arterial relaxation by N(ε)-lysine acetylation of contractile proteins. Arterioscler Thromb Vasc Biol 2012; 32:2435-43. [PMID: 22859492 DOI: 10.1161/atvbaha.112.254011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To address the role of epigenetic enzymes in the process of arterial vasorelaxation and nitrate tolerance, in vitro and in vivo experiments were performed in the presence or absence of glyceryl trinitrate (GTN) or histone deacetylases/histone acetylases modulators. METHODS AND RESULTS In vitro single GTN administration rapidly increased cGMP synthesis and protein N(ε)-lysine acetylation in rat smooth muscle cells, including myosin light chain and smooth muscle actin. This phenomenon determined a decrease in myosin light chain phosphorylation and actomyosin formation. These effects were abolished by prolonged exposure to GTN and rescued by treatment with trichostatin A. In vivo, adult male rats were treated for 72 hours with subcutaneous injections of GTN alone or in combination with the histone deacetylases inhibitors trichostatin A, suberoylanilide hydroxamic acid, MS-27-275, or valproic acid. Ex vivo experiments performed on aortic rings showed that the effect of tolerance was reversed by all proacetylation drugs, including the p300/CREB binding protein-associated factor activator pentadecylidenemalonate 1b (SPV106). Any response to GTN was abolished by anacardic acid, a potent histone acetylases inhibitor. CONCLUSIONS This study establishes the following points: (1) GTN treatment increases histone acetylases activity; (2) GTN-activated p300/CREB binding protein-associated factor increases protein N(ε)-lysine acetylation; (3) N(ε)-lysine acetylation of contractile proteins influences GTN-dependent vascular response. Hence, combination of epigenetic drugs and nitroglycerin may be envisaged as a novel treatment strategy for coronary artery disease symptoms and other cardiovascular accidents of ischemic origin.
Collapse
Affiliation(s)
- Claudia Colussi
- Laboratorio di PatologiaVascolare, Istituto Dermopatico dell’Immacolata, Roma, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Yuan Q, Tang W, Zhang X, Hinson JA, Liu C, Osei K, Wang J. Proinsulin atypical maturation and disposal induces extensive defects in mouse Ins2+/Akita β-cells. PLoS One 2012; 7:e35098. [PMID: 22509386 PMCID: PMC3318013 DOI: 10.1371/journal.pone.0035098] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 03/08/2012] [Indexed: 02/06/2023] Open
Abstract
Because of its low relative folding rate and plentiful manufacture in β-cells, proinsulin maintains a homeostatic balance of natively and plentiful non-natively folded states (i.e., proinsulin homeostasis, PIHO) through the integration of maturation and disposal processes. PIHO is susceptible to genetic and environmental influences, and its disorder has been critically linked to defects in β-cells in diabetes. To explore this hypothesis, we performed polymerase chain reaction (PCR), metabolic-labeling, immunoblotting, and histological studies to clarify what defects result from primary disorder of PIHO in model Ins2+/Akita β-cells. We used T antigen-transformed Ins2+/Akita and control Ins2+/+ β-cells established from Akita and wild-type littermate mice. In Ins2+/Akita β-cells, we found no apparent defect at the transcriptional and translational levels to contribute to reduced cellular content of insulin and its precursor and secreted insulin. Glucose response remained normal in proinsulin biosynthesis but was impaired for insulin secretion. The size and number of mature insulin granules were reduced, but the size/number of endoplasmic reticulum, Golgi, mitochondrion, and lysosome organelles and vacuoles were expanded/increased. Moreover, cell death increased, and severe oxidative stress, which manifested as increased reactive oxygen species, thioredoxin-interacting protein, and protein tyrosine nitration, occurred in Ins2+/Akita β-cells and/or islets. These data show the first clear evidence that primary PIHO imbalance induces severe oxidative stress and impairs glucose-stimulated insulin release and β-cell survival as well as producing other toxic consequences. The defects disclosed/clarified in model Ins2+/Akita β-cells further support a role of the genetic and stress-susceptible PIHO disorder in β-cell failure and diabetes.
Collapse
Affiliation(s)
- Qingxin Yuan
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Wei Tang
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Xiaoping Zhang
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Jack A. Hinson
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Chao Liu
- Division of Endocrinology, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese and Western Medicine, Nanjing, Jiangsu, China
| | - Kwame Osei
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Jie Wang
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Division of Endocrinology, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese and Western Medicine, Nanjing, Jiangsu, China
- * E-mail:
| |
Collapse
|
46
|
Bedoya FJ, Salguero-Aranda C, Cahuana GM, Tapia-Limonchi R, Soria B, Tejedo JR. Regulation of pancreatic β-cell survival by nitric oxide: clinical relevance. Islets 2012; 4:108-18. [PMID: 22614339 DOI: 10.4161/isl.19822] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The reduction of pancreatic β-cell mass is an important factor in the development of type 1 and type 2 diabetes. Understanding the mechanisms that regulate the maintenance of pancreatic β-cell mass as well as β-cell death is necessary for the establishment of therapeutic strategies. In this context, nitric oxide (NO) is a diatomic, gaseous, highly reactive molecule with biological activity that participates in the regulation of pancreatic β-cell mass. Two types of cellular responses can be distinguished depending on the level of NO production. First, pancreatic β-cells exposed to inflammatory cytokines, lipid stress or hyperglycaemia produce high concentrations of NO, mainly due to the activation of inducible NO synthase (iNOS), thus promoting cell death. Meanwhile, under homeostatic conditions, low concentrations of NO, constitutively produced by endothelial NO synthase (eNOS), promote cell survival. Here, we will discuss the current knowledge of the NO-dependent mechanisms activated during cellular responses, emphasizing those related to the regulation of cell survival.
Collapse
Affiliation(s)
- Francisco J Bedoya
- Andalusian Center for Molecular Biology and Regenerative Medicine, University Pablo de Olavide, CIBERDEM, RED-TERCEL, Seville, Spain
| | | | | | | | | | | |
Collapse
|
47
|
Hess DT, Stamler JS. Regulation by S-nitrosylation of protein post-translational modification. J Biol Chem 2011; 287:4411-8. [PMID: 22147701 DOI: 10.1074/jbc.r111.285742] [Citation(s) in RCA: 296] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Protein post-translational modification by S-nitrosylation conveys a ubiquitous influence of nitric oxide on signal transduction in eukaryotic cells. The wide functional purview of S-nitrosylation reflects in part the regulation by S-nitrosylation of the principal protein post-translational modifications that play a role in cell signaling, including phosphorylation, acetylation, ubiquitylation and related modifications, palmitoylation, and alternative Cys-based redox modifications. In this minireview, we discuss the mechanisms through which S-nitrosylation exerts its broad pleiotropic influence on protein post-translational modification.
Collapse
Affiliation(s)
- Douglas T Hess
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | |
Collapse
|
48
|
Krause B, Hanson M, Casanello P. Role of nitric oxide in placental vascular development and function. Placenta 2011; 32:797-805. [PMID: 21798594 PMCID: PMC3218217 DOI: 10.1016/j.placenta.2011.06.025] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 06/28/2011] [Accepted: 06/29/2011] [Indexed: 11/27/2022]
Abstract
Nitric oxide (NO) is one of the most pleiotropic signaling molecules at systemic and cellular levels, participating in vascular tone regulation, cellular respiration, proliferation, apoptosis and gene expression. Indeed NO actively participates in trophoblast invasion, placental development and represents the main vasodilator in this tissue. Despite the large number of studies addressing the role of NO in the placenta, its participation in placental vascular development and the effect of altered levels of NO on placental function remains to be clarified. This review draws a time-line of the participation of NO throughout placental vascular development, from the differentiation of vascular precursors to the consolidation of vascular function are considered. The influence of NO on cell types involved in the origin of the placental vasculature and the expression and function of the nitric oxide synthases (NOS) throughout pregnancy are described. The developmental processes involved in the placental vascular bed are considered, such as the participation of NO in placental vasculogenesis and angiogenesis through VEGF and Angiopoietin signaling molecules. The role of NO in vascular function once the placental vascular tree has developed, in normal pregnancy as well as in pregnancy-related diseases, is then discussed.
Collapse
Affiliation(s)
- B.J. Krause
- Division of Obstetrics and Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - M.A. Hanson
- Institute of Developmental Sciences, Academic Unit of Human Development & Health, Faculty of Medicine, University of Southampton, SO16 6YD, UK
| | - P. Casanello
- Division of Obstetrics and Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| |
Collapse
|
49
|
Wang HH, Hsieh HL, Yang CM. Nitric oxide production by endothelin-1 enhances astrocytic migration via the tyrosine nitration of matrix metalloproteinase-9. J Cell Physiol 2011; 226:2244-56. [PMID: 21660948 DOI: 10.1002/jcp.22560] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The deleterious effects of endothelin-1 (ET-1) in the central nervous system (CNS) include disturbance of water homeostasis and blood-brain barrier (BBB) integrity. In the CNS, ischemic injury elicits ET-1 release from astrocytes, behaving through G-protein coupled ET receptors. These considerations raise the question of whether ET-1 influences cellular functions of astrocytes, the major cell type that provides structural and functional support for neurons. Uncontrolled nitric oxide (NO) production has been implicated in sterile brain insults, neuroinflammation, and neurodegenerative diseases, which involve astrocyte activation and neuronal death. However, the detailed mechanisms of ET-1 action related to NO release on rat brain astrocytes (RBA-1) remain unknown. In this study, we demonstrate that exposure of astrocytes to ET-1 results in the inducible nitric oxide synthase (iNOS) up-regulation, NO production, and matrix metalloproteinase-9 (MMP-9) activation in astrocytes. The data obtained with Western blot, reverse transcription-PCR (RT-PCR), and immunofluorescent staining analyses showed that ET-1-induced iNOS expression and NO production were mediated through an ET(B)-dependent transcriptional activation. Engagement of G(i/o)--and G(q) -coupled ET(B) receptors by ET-1 led to activation of c-Src-dependent phosphoinositide 3-kinase (PI3K)/Akt and p42/p44 mitogen-activated protein kinase (MAPK) and then activated transcription factor nuclear factor-κB (NF-κB). The activated NF-κB was translocated into nucleus and thereby promoted iNOS gene transcription. Ultimately, NO production stimulated by ET-1 enhanced the migration of astrocytes through the tyrosine nitration of MMP-9. Taken together, these results suggested that in astrocytes, activation of NF-κB by ET(B)-dependent c-Src, PI3K/Akt, and p42/p44 MAPK signalings is necessary for ET-1-induced iNOS gene up-regulation.
Collapse
Affiliation(s)
- Hui-Hsin Wang
- Department of Pharmacology, Chang Gung University, Tao-Yuan, Taiwan
| | | | | |
Collapse
|
50
|
Affiliation(s)
- Diane E. Handy
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Rita Castro
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Metabolism & Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon, Portugal
| | - Joseph Loscalzo
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|