251
|
Miguel-Hidalgo JJ. Astroglia in the Vulnerability and Maintenance of Alcohol Use Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:255-279. [PMID: 34888838 DOI: 10.1007/978-3-030-77375-5_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Changes induced in the morphology and the multiplicity of functional roles played by astrocytes in brain regions critical to the establishment and maintenance of alcohol abuse suggest that they make an important contribution to the vulnerability to alcohol use disorders. The understanding of the relevant mechanisms accounting for that contribution is complicated by the fact that alcohol itself acts directly on astrocytes altering their metabolism, gene expression, and plasticity, so that the ultimate result is a complex interaction of various cellular pathways, including intracellular calcium regulation, neuroimmune responses, and regulation of neurotransmitter and gliotransmitter release and uptake. The recent years have seen a steady increase in the characterization of several of the relevant mechanisms, but much remains to be done for a full understanding of the astrocytes' contribution to the vulnerability to alcohol dependence and abuse and for using that knowledge in designing effective therapies for AUDs.
Collapse
Affiliation(s)
- José Javier Miguel-Hidalgo
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
252
|
Luhar I, Luhar S, Abdullah MMAB, Razak RA, Vizureanu P, Sandu AV, Matasaru PD. A State-of-the-Art Review on Innovative Geopolymer Composites Designed for Water and Wastewater Treatment. MATERIALS (BASEL, SWITZERLAND) 2021; 14:7456. [PMID: 34885611 PMCID: PMC8658912 DOI: 10.3390/ma14237456] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/17/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022]
Abstract
There is nothing more fundamental than clean potable water for living beings next to air. On the other hand, wastewater management is cropping up as a challenging task day-by-day due to lots of new additions of novel pollutants as well as the development of infrastructures and regulations that could not maintain its pace with the burgeoning escalation of populace and urbanizations. Therefore, momentous approaches must be sought-after to reclaim fresh water from wastewaters in order to address this great societal challenge. One of the routes is to clean wastewater through treatment processes using diverse adsorbents. However, most of them are unsustainable and quite costly e.g. activated carbon adsorbents, etc. Quite recently, innovative, sustainable, durable, affordable, user and eco-benevolent Geopolymer composites have been brought into play to serve the purpose as a pretty novel subject matter since they can be manufactured by a simple process of Geopolymerization at low temperature, lower energy with mitigated carbon footprints and marvellously, exhibit outstanding properties of physical and chemical stability, ion-exchange, dielectric characteristics, etc., with a porous structure and of course lucrative too because of the incorporation of wastes with them, which is in harmony with the goal to transit from linear to circular economy, i.e., "one's waste is the treasure for another". For these reasons, nowadays, this ground-breaking inorganic class of amorphous alumina-silicate materials are drawing the attention of the world researchers for designing them as adsorbents for water and wastewater treatment where the chemical nature and structure of the materials have a great impact on their adsorption competence. The aim of the current most recent state-of-the-art and scientometric review is to comprehend and assess thoroughly the advancements in geo-synthesis, properties and applications of geopolymer composites designed for the elimination of hazardous contaminants viz., heavy metal ions, dyes, etc. The adsorption mechanisms and effects of various environmental conditions on adsorption efficiency are also taken into account for review of the importance of Geopolymers as most recent adsorbents to get rid of the death-defying and toxic pollutants from wastewater with a view to obtaining reclaimed potable and sparkling water for reuse offering to trim down the massive crisis of scarcity of water promoting sustainable water and wastewater treatment for greener environments. The appraisal is made on the performance estimation of Geopolymers for water and wastewater treatment along with the three-dimensional printed components are characterized for mechanical, physical and chemical attributes, permeability and Ammonium (NH4+) ion removal competence of Geopolymer composites as alternative adsorbents for sequestration of an assortment of contaminants during wastewater treatment.
Collapse
Affiliation(s)
- Ismail Luhar
- Department of Civil Engineering, Shri Jagdishprasad Jhabarmal Tibrewala University, Rajasthan 333001, India;
| | - Salmabanu Luhar
- Center of Excellence Geopolymer and Green Technology (CEGeoGTech), Universiti Malaysia Perlis (UniMAP), Perlis 01000, Malaysia;
- Frederick Research Center, P.O. Box 24729, Nicosia 1303, Cyprus
- Department of Civil Engineering, Frederick University, Nicosia 1036, Cyprus
| | - Mohd Mustafa Al Bakri Abdullah
- Center of Excellence Geopolymer and Green Technology (CEGeoGTech), Universiti Malaysia Perlis (UniMAP), Perlis 01000, Malaysia;
| | - Rafiza Abdul Razak
- Center of Excellence Geopolymer and Green Technology (CEGeoGTech), Universiti Malaysia Perlis (UniMAP), Perlis 01000, Malaysia;
| | - Petrica Vizureanu
- Faculty of Materials Science and Engineering, Gheorghe Asachi Technical University of Iasi, D. Mangeron 41, 700050 Iasi, Romania
| | - Andrei Victor Sandu
- Faculty of Materials Science and Engineering, Gheorghe Asachi Technical University of Iasi, D. Mangeron 41, 700050 Iasi, Romania
- Romanian Inventors Forum, St. P. Movila 3, 700089 Iasi, Romania
- National Institute for Research and Development in Environmental Protection INCDPM, Splaiul Independentei 294, 060031 Bucuresti, Romania
| | - Petre-Daniel Matasaru
- Faculty of Electronics, Telecommunications and Information Technology, Technical University “Gheorghe Asachi”, Carol I Bvd, nr. 11 A, 700506 Iasi, Romania;
| |
Collapse
|
253
|
Karanikas E. Psychologically Traumatic Oxidative Stress; A Comprehensive Review of Redox Mechanisms and Related Inflammatory Implications. PSYCHOPHARMACOLOGY BULLETIN 2021; 51:65-86. [PMID: 34887600 PMCID: PMC8601764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The organism's energy requirements for homeostatic balance are covered by the redox mechanisms. Yet in case of psychologically traumatic stress, allostatic regulations activate both pro-oxidant and antioxidant molecules as well as respective components of the inflammatory system. Thus a new setpoint of dynamic interactions among redox elements is reached. Similarly, a multifaceted interplay between redox and inflammatory fields is activated with the mediation of major effector systems such as the immune system, Hypothalamic-Pituitary-Adrenal axis, kynurenine, and the glycaemic regulatory one. In case of sustained and/or intense traumatic stress the prophylactic antioxidant components are inadequate to provide the organism with neuroprotection finally culminating in Oxidative Stress and subsequently to cellular apoptosis. In parallel multiple inflammatory systems trigger and/or are triggered by the redox systems in tight fashion so that the causation sequence appears obscure. This exhaustive review aims at the comprehension of the interaction among components of the redox system as well as to the collection of disperse findings relative to the redox-inflammatory interplay in the context of traumatic stress so that new research strategies could be developed.
Collapse
Affiliation(s)
- Evangelos Karanikas
- Karanikas, Department of Psychiatry, General Military Hospital, Thessaloniki, Greece
| |
Collapse
|
254
|
Al-Mubarak BR, Bell KFS, Chowdhry S, Meakin PJ, Baxter PS, McKay S, Dando O, Ashford MLJ, Gazaryan I, Hayes JD, Hardingham GE. Non-canonical Keap1-independent activation of Nrf2 in astrocytes by mild oxidative stress. Redox Biol 2021; 47:102158. [PMID: 34626892 PMCID: PMC8512624 DOI: 10.1016/j.redox.2021.102158] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/30/2022] Open
Abstract
The transcription factor Nrf2 is a stress-responsive master regulator of antioxidant, detoxification and proteostasis genes. In astrocytes, Nrf2-dependent gene expression drives cell-autonomous cytoprotection and also non-cell-autonomous protection of nearby neurons, and can ameliorate pathology in several acute and chronic neurological disorders associated with oxidative stress. However, the value of astrocytic Nrf2 as a therapeutic target depends in part on whether Nrf2 activation by disease-associated oxidative stress occludes the effect of any Nrf2-activating drug. Nrf2 activation classically involves the inhibition of interactions between Nrf2's Neh2 domain and Keap1, which directs Nrf2 degradation. Keap1 inhibition is mediated by the modification of cysteine residues on Keap1, and can be triggered by electrophilic small molecules such as tBHQ. Here we show that astrocytic Nrf2 activation by oxidative stress involves Keap1-independent non-canonical signaling. Keap1 deficiency elevates basal Nrf2 target gene expression in astrocytes and occludes the effects of tBHQ, oxidative stress still induced strong Nrf2-dependent gene expression in Keap1-deficient astrocytes. Moreover, while tBHQ prevented protein degradation mediated via Nrf2's Neh2 domain, oxidative stress did not, consistent with a Keap1-independent mechanism. Moreover the effects of oxidative stress and tBHQ on Nrf2 target gene expression are additive, not occlusive. Mechanistically, oxidative stress enhances the transactivation potential of Nrf2's Neh5 domain in a manner dependent on its Cys-191 residue. Thus, astrocytic Nrf2 activation by oxidative stress involves Keap1-independent non-canonical signaling, meaning that further Nrf2 activation by Keap1-inhibiting drugs may be a viable therapeutic strategy.
Collapse
Affiliation(s)
- Bashayer R Al-Mubarak
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK; Behavioral Genetics Unit, Department of Genetics, King Faisal Specialist Hospital and Research Center, P.O Box 3354, Riyadh, 11211, Saudi Arabia
| | - Karen F S Bell
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Sudhir Chowdhry
- Biomedical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Paul J Meakin
- Discovery & Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, LS2 9JT, UK; Division of Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Paul S Baxter
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, EH16 4SB, UK
| | - Sean McKay
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, EH16 4SB, UK
| | - Owen Dando
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, EH16 4SB, UK
| | - Michael L J Ashford
- Division of Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Irina Gazaryan
- Department of Chemistry and Physical Sciences, Dyson College of Arts and Sciences, Pace University, Pleasantville, NY, 10570, USA
| | - John D Hayes
- Biomedical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, EH16 4SB, UK.
| |
Collapse
|
255
|
Schultz B, Taday J, Menezes L, Cigerce A, Leite MC, Gonçalves CA. Calpain-Mediated Alterations in Astrocytes Before and During Amyloid Chaos in Alzheimer's Disease. J Alzheimers Dis 2021; 84:1415-1430. [PMID: 34719501 DOI: 10.3233/jad-215182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
One of the changes found in the brain in Alzheimer's disease (AD) is increased calpain, derived from calcium dysregulation, oxidative stress, and/or neuroinflammation, which are all assumed to be basic pillars in neurodegenerative diseases. The role of calpain in synaptic plasticity, neuronal death, and AD has been discussed in some reviews. However, astrocytic calpain changes sometimes appear to be secondary and consequent to neuronal damage in AD. Herein, we explore the possibility of calpain-mediated astroglial reactivity in AD, both preceding and during the amyloid phase. We discuss the types of brain calpains but focus the review on calpains 1 and 2 and some important targets in astrocytes. We address the signaling involved in controlling calpain expression, mainly involving p38/mitogen-activated protein kinase and calcineurin, as well as how calpain regulates the expression of proteins involved in astroglial reactivity through calcineurin and cyclin-dependent kinase 5. Throughout the text, we have tried to provide evidence of the connection between the alterations caused by calpain and the metabolic changes associated with AD. In addition, we discuss the possibility that calpain mediates amyloid-β clearance in astrocytes, as opposed to amyloid-β accumulation in neurons.
Collapse
Affiliation(s)
- Bruna Schultz
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jéssica Taday
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Leonardo Menezes
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Anderson Cigerce
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marina C Leite
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Carlos-Alberto Gonçalves
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
256
|
Tobeiha M, Rajabi A, Raisi A, Mohajeri M, Yazdi SM, Davoodvandi A, Aslanbeigi F, Vaziri M, Hamblin MR, Mirzaei H. Potential of natural products in osteosarcoma treatment: Focus on molecular mechanisms. Biomed Pharmacother 2021; 144:112257. [PMID: 34688081 DOI: 10.1016/j.biopha.2021.112257] [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: 08/03/2021] [Revised: 09/21/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is the most frequent type of bone cancer found in children and adolescents, and commonly arises in the metaphyseal region of tubular long bones. Standard therapeutic approaches, such as surgery, chemotherapy, and radiation therapy, are used in the management of osteosarcoma. In recent years, the mortality rate of osteosarcoma has decreased due to advances in treatment methods. Today, the scientific community is investigating the use of different naturally derived active principles against various types of cancer. Natural bioactive compounds can function against cancer cells in two ways. Firstly they can act as classical cytotoxic compounds by non-specifically affecting macromolecules, such as DNA, enzymes, and microtubules, which are also expressed in normal proliferating cells, but to a greater extent by cancer cells. Secondly, they can act against oncogenic signal transduction pathways, many of which are activated in cancer cells. Some bioactive plant-derived agents are gaining increasing attention because of their anti-cancer properties. Moreover, some naturally-derived compounds can significantly promote the effectiveness of standard chemotherapy drugs, and in certain cases are able to ameliorate drug-induced adverse effects caused by chemotherapy. In the present review we summarize the effects of various naturally-occurring bioactive compounds against osteosarcoma.
Collapse
Affiliation(s)
- Mohammad Tobeiha
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Rajabi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Arash Raisi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahshad Mohajeri
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Amirhossein Davoodvandi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Aslanbeigi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - MohamadSadegh Vaziri
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
257
|
Abstract
Protein oxidation is a topic of indisputable scientific interest given the impact of oxidized proteins on food quality and safety. Carbonylation is regarded as one of the most notable post-translational modifications in proteins and yet, this reaction and its consequences are poorly understood. From a mechanistic perspective, primary protein carbonyls (i.e. α-aminoadipic and γ-glutamic semialdehydes) have been linked to radical-mediated oxidative stress, but recent studies emphasize the role alternative carbonylation pathways linked to the Maillard reaction. Secondary protein carbonyls are introduced in proteins via covalent linkage of lipid carbonyls (i.e. protein-bound malondialdehyde). The high reactivity of protein carbonyls in foods and other biological systems indicates the intricate chemistry of these species and urges further research to provide insight into these molecular mechanisms and pathways. In particular, protein carbonyls are involved in the formation of aberrant and dysfunctional protein aggregates, undergo further oxidation to yield carboxylic acids of biological relevance and establish interactions with other biomolecules such as oxidizing lipids and phytochemicals. From a methodological perspective, the routine dinitrophenylhydrazine (DNPH) method is criticized not only for the lack of accuracy and consistency but also authors typically perform a poor interpretation of DNPH results, which leads to misleading conclusions. From a practical perspective, the biological relevance of protein carbonyls in the field of food science and nutrition is still a topic of debate. Though the implication of carbonylation on impaired protein functionality and poor protein digestibility is generally recognized, the underlying mechanism of such connections requires further clarification. From a medical perspective, protein carbonyls are highlighted as markers of protein oxidation, oxidative stress and disease. Yet, the specific role of specific protein carbonyls in the onset of particular biological impairments needs further investigations. Recent studies indicates that regardless of the origin (in vivo or dietary) protein carbonyls may act as signalling molecules which activate not only the endogenous antioxidant defences but also implicate the immune system. The present paper concisely reviews the most recent advances in this topic to identify, when applicable, potential fields of interest for future studies.
Collapse
|
258
|
Padilla P, Andrade MJ, Peña FJ, Rodríguez A, Estévez M. An in vitro assay of the effect of lysine oxidation end-product, α-aminoadipic acid, on the redox status and gene expression in probiotic Lactobacillus reuteri PL503. Amino Acids 2021; 54:663-673. [PMID: 34657206 PMCID: PMC9117375 DOI: 10.1007/s00726-021-03087-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/04/2021] [Indexed: 01/18/2023]
Abstract
This study was designed to gain information about the underlying mechanisms of the effects of a food-occurring free oxidized amino acid, α-aminoadipic acid (AAA), on the probiotic Lactobacillus reuteri PL503. This bacterium was incubated in colonic-simulated conditions (37 °C for 24 h in microaerophilic conditions) and exposed to three food-compatible AAA concentrations, namely, 1 mM, 5 mM, and 10 mM. A control group with no AAA exposure was also considered. Each of the four experimental conditions was replicated three times and samplings were collected at 12, 16, 20, and 24 h. The downregulation of the uspA gene by AAA (0.5-fold decrease as compared to control) suggests that AAA is identified as a potential chemical threat. The dhaT gene, implicated in the antioxidant defense, was found to be upregulated in bacteria treated with 1 and 5 mM AAA (up to twofold increase, as compared to control), which suggest the ability of the oxidized amino acid to impair the redox status of the bacterium. In fact, AAA caused an increased production of reactive oxygen species (ROS) and the accretion of post-translational changes (protein carbonylation) in L. reuteri (up to 13 nmol allysine/mg protein vs 1.8 nmol allysine/mg protein in control). These results suggest that probiotic bacteria identify oxidized amino acids as harmful species and activate mechanisms that may protect themselves and the host against their noxious effects.
Collapse
Affiliation(s)
- Patricia Padilla
- Food Technology, IPROCAR Research Institute, University of Extremadura, 10003, Cáceres, Spain.,Faculty of Veterinary Science, IPROCAR Research Institute, Food Hygiene and Safety, University of Extremadura, 10003, Cáceres, Spain
| | - María J Andrade
- Faculty of Veterinary Science, IPROCAR Research Institute, Food Hygiene and Safety, University of Extremadura, 10003, Cáceres, Spain
| | - Fernando J Peña
- Laboratory of Equine Reproduction and Equine Spermatology, University of Extremadura, 10003, Cáceres, Spain
| | - Alicia Rodríguez
- Faculty of Veterinary Science, IPROCAR Research Institute, Food Hygiene and Safety, University of Extremadura, 10003, Cáceres, Spain
| | - Mario Estévez
- Food Technology, IPROCAR Research Institute, University of Extremadura, 10003, Cáceres, Spain.
| |
Collapse
|
259
|
Tian X, Zhang S, Zhou L, Seyhan AA, Hernandez Borrero L, Zhang Y, El-Deiry WS. Targeting the Integrated Stress Response in Cancer Therapy. Front Pharmacol 2021; 12:747837. [PMID: 34630117 PMCID: PMC8498116 DOI: 10.3389/fphar.2021.747837] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/10/2021] [Indexed: 12/11/2022] Open
Abstract
The integrated stress response (ISR) is an evolutionarily conserved intra-cellular signaling network which is activated in response to intrinsic and extrinsic stresses. Various stresses are sensed by four specialized kinases, PKR-like ER kinase (PERK), general control non-derepressible 2 (GCN2), double-stranded RNA-dependent protein kinase (PKR) and heme-regulated eIF2α kinase (HRI) that converge on phosphorylation of serine 51 of eIF2α. eIF2α phosphorylation causes a global reduction of protein synthesis and triggers the translation of specific mRNAs, including activating transcription factor 4 (ATF4). Although the ISR promotes cell survival and homeostasis, when stress is severe or prolonged the ISR signaling will shift to regulate cellular apoptosis. We review the ISR signaling pathway, regulation and importance in cancer therapy.
Collapse
Affiliation(s)
- Xiaobing Tian
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States.,Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States.,Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States.,Cancer Center at Brown University, Providence, RI, United States
| | - Shengliang Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States.,Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States.,Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States.,Cancer Center at Brown University, Providence, RI, United States
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States.,Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States.,Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States.,Cancer Center at Brown University, Providence, RI, United States
| | - Attila A Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States.,Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States.,Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States.,Cancer Center at Brown University, Providence, RI, United States
| | - Liz Hernandez Borrero
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Yiqun Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States.,Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States.,Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States.,Cancer Center at Brown University, Providence, RI, United States.,Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, RI, United States
| |
Collapse
|
260
|
Cuadrado A. Brain-Protective Mechanisms of Transcription Factor NRF2: Toward a Common Strategy for Neurodegenerative Diseases. Annu Rev Pharmacol Toxicol 2021; 62:255-277. [PMID: 34637322 DOI: 10.1146/annurev-pharmtox-052220-103416] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neurodegenerative diseases are characterized by the loss of homeostatic functions that control redox and energy metabolism, neuroinflammation, and proteostasis. The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) is a master controller of these functions, and its overall activity is compromised during aging and in these diseases. However, NRF2 can be activated pharmacologically and is now being considered a common therapeutic target. Many gaps still exist in our knowledge of the specific role that NRF2 plays in specialized brain cell functions or how these cells respond to the hallmarks of these diseases. This review discusses the relevance of NRF2 to several hallmark features of neurodegenerative diseases and the current status of pharmacological activators that might pass through the blood-brain barrier and provide a disease-modifying effect. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid, Madrid 28049, Spain.,Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid 28029, Spain.,Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid 28046, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid 28031, Spain;
| |
Collapse
|
261
|
Bastani S, Akbarzadeh M, Rastgar Rezaei Y, Farzane A, Nouri M, Mollapour Sisakht M, Fattahi A, Akbarzadeh M, Reiter RJ. Melatonin as a Therapeutic Agent for the Inhibition of Hypoxia-Induced Tumor Progression: A Description of Possible Mechanisms Involved. Int J Mol Sci 2021; 22:10874. [PMID: 34639215 PMCID: PMC8509383 DOI: 10.3390/ijms221910874] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 12/27/2022] Open
Abstract
Hypoxia has an important role in tumor progression via the up-regulation of growth factors and cellular adaptation genes. These changes promote cell survival, proliferation, invasion, metastasis, angiogenesis, and energy metabolism in favor of cancer development. Hypoxia also plays a central role in determining the resistance of tumors to chemotherapy. Hypoxia of the tumor microenvironment provides an opportunity to develop new therapeutic strategies that may selectively induce apoptosis of the hypoxic cancer cells. Melatonin is well known for its role in the regulation of circadian rhythms and seasonal reproduction. Numerous studies have also documented the anti-cancer properties of melatonin, including anti-proliferation, anti-angiogenesis, and apoptosis promotion. In this paper, we hypothesized that melatonin exerts anti-cancer effects by inhibiting hypoxia-induced pathways. Considering this action, co-administration of melatonin in combination with other therapeutic medications might increase the effectiveness of anti-cancer drugs. In this review, we discussed the possible signaling pathways by which melatonin inhibits hypoxia-induced cancer cell survival, invasion, migration, and metabolism, as well as tumor angiogenesis.
Collapse
Affiliation(s)
- Sepideh Bastani
- Research Center for Pharmaceutical Nanotechnology (RCPN), Tabriz University of Medical Sciences, Tabriz 51368, Iran;
- Stem Cell And Regenerative Medicine Institute (SCARM), Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Moloud Akbarzadeh
- Stem Cell And Regenerative Medicine Institute (SCARM), Tabriz University of Medical Sciences, Tabriz 51368, Iran;
- Department of Cellular and Molecular Biology, Faculty of Biological Science, Azarbaijan Shahid Madani University, Tabriz 51368, Iran
| | - Yeganeh Rastgar Rezaei
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Ali Farzane
- Department of Health Information Management, School of Allied Medical Science, Tehran University of Medical Sciences, Tehran 11369, Iran;
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Mahsa Mollapour Sisakht
- Stem Cell and Regenerative Medicine Center of Excellence, Tehran University of Medical Sciences, Tehran 11369, Iran;
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen–Nürnberg, Comprehensive Cancer Center ER-EMN, 91054 Erlangen, Germany
| | - Maryam Akbarzadeh
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health, Long School of Medicine, San Antonio, TX 78229, USA;
| |
Collapse
|
262
|
Kukula-Koch W, Szwajgier D, Gaweł-Bęben K, Strzępek-Gomółka M, Głowniak K, Meissner HO. Is Phytomelatonin Complex Better Than Synthetic Melatonin? The Assessment of the Antiradical and Anti-Inflammatory Properties. Molecules 2021; 26:6087. [PMID: 34641628 PMCID: PMC8512846 DOI: 10.3390/molecules26196087] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022] Open
Abstract
This work aims to assess the recently established anti-inflammatory and antioxidant potential of melatonin of plant origin extracted from the plant matrix as a phytomelatonin complex (PHT-MLT), and compare its activity with synthetic melatonin (SNT-MLT) when used on its own or with vitamin C. For this purpose, a COX-2 enzyme inhibitory activity test, an antiradical activity in vitro and on cell lines assays, was performed on both PHT-MLT and SNT-MLT products. COX-2 inhibitory activity of PHT-MLT was found to be ca. 6.5 times stronger than that of SNT-MLT (43.3% and 6.7% enzyme inhibition, equivalent to the activity of acetylsalicylic acid in conc. 30.3 ± 0.2 and 12.0 ± 0.3 mg/mL, respectively). Higher antiradical potential and COX-2 inhibitory properties of PHT-MLT could be explained by the presence of additional naturally occurring constituents in alfalfa, chlorella, and rice, which were clearly visible on the HPLC-ESI-QTOF-MS fingerprint. The antiradical properties of PHT-MLT determined in the DPPH test (IC50 of 21.6 ± 1 mg of powder/mL) were found to originate from the presence of other metabolites in the 50% EtOH extract while SNT-MLT was found to be inactive under the applied testing conditions. However, the antioxidant studies on HaCaT keratinocytes stimulated with H2O2 revealed a noticeable activity in all samples. The presence of PHT-MLT (12.5, 25 and 50 µg/mL) and vitamin C (12.5, 25 and 50 µg/mL) in the H2O2-pretreated HaCaT keratinocytes protected the cells from generating reactive oxygen species. This observation confirms that MLT-containing samples affect the intracellular production of enzymes and neutralize the free radicals. Presented results indicated that MLT-containing products in combination with Vitamin C dosage are worth to be considered as a preventive alternative in the therapy of various diseases in the etiopathogenesis, of which radical and inflammatory mechanisms play an important role.
Collapse
Affiliation(s)
- Wirginia Kukula-Koch
- Department of Pharmacognosy with Garden of Medicinal Plants, Medicinal University in Lublin, 1 Chodźki Str., 20-093 Lublin, Poland
| | - Dominik Szwajgier
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences, 8 Skromna Str., 20-704 Lublin, Poland;
| | - Katarzyna Gaweł-Bęben
- Department of Cosmetology, University of Information Technology and Management in Rzeszów, 2 Sucharskiego Str., 35-225 Rzeszów, Poland; (K.G.-B.); (M.S.-G.); (K.G.)
| | - Marcelina Strzępek-Gomółka
- Department of Cosmetology, University of Information Technology and Management in Rzeszów, 2 Sucharskiego Str., 35-225 Rzeszów, Poland; (K.G.-B.); (M.S.-G.); (K.G.)
| | - Kazimierz Głowniak
- Department of Cosmetology, University of Information Technology and Management in Rzeszów, 2 Sucharskiego Str., 35-225 Rzeszów, Poland; (K.G.-B.); (M.S.-G.); (K.G.)
| | - Henry O. Meissner
- Therapeutic Research, TTD International Pty Ltd., 39 Leopard Ave., Gold Coast 4221, Australia;
| |
Collapse
|
263
|
Ahmadpour N, Kantroo M, Stobart JL. Extracellular Calcium Influx Pathways in Astrocyte Calcium Microdomain Physiology. Biomolecules 2021; 11:1467. [PMID: 34680100 PMCID: PMC8533159 DOI: 10.3390/biom11101467] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/25/2021] [Accepted: 10/01/2021] [Indexed: 02/08/2023] Open
Abstract
Astrocytes are complex glial cells that play many essential roles in the brain, including the fine-tuning of synaptic activity and blood flow. These roles are linked to fluctuations in intracellular Ca2+ within astrocytes. Recent advances in imaging techniques have identified localized Ca2+ transients within the fine processes of the astrocytic structure, which we term microdomain Ca2+ events. These Ca2+ transients are very diverse and occur under different conditions, including in the presence or absence of surrounding circuit activity. This complexity suggests that different signalling mechanisms mediate microdomain events which may then encode specific astrocyte functions from the modulation of synapses up to brain circuits and behaviour. Several recent studies have shown that a subset of astrocyte microdomain Ca2+ events occur rapidly following local neuronal circuit activity. In this review, we consider the physiological relevance of microdomain astrocyte Ca2+ signalling within brain circuits and outline possible pathways of extracellular Ca2+ influx through ionotropic receptors and other Ca2+ ion channels, which may contribute to astrocyte microdomain events with potentially fast dynamics.
Collapse
Affiliation(s)
| | | | - Jillian L. Stobart
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, 750 McDermot Avenue, Winnipeg, MG R3E 0T5, Canada; (N.A.); (M.K.)
| |
Collapse
|
264
|
NUPR1 inhibitor ZZW-115 induces ferroptosis in a mitochondria-dependent manner. Cell Death Discov 2021; 7:269. [PMID: 34599149 PMCID: PMC8486797 DOI: 10.1038/s41420-021-00662-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/20/2021] [Accepted: 09/07/2021] [Indexed: 12/18/2022] Open
Abstract
Ferroptosis is an iron-dependent cell death characterized by the accumulation of hydroperoxided phospholipids. Here, we report that the NUPR1 inhibitor ZZW-115 induces ROS accumulation followed by a ferroptotic cell death, which could be prevented by ferrostatin-1 (Fer-1) and ROS-scavenging agents. The ferroptotic activity can be improved by inhibiting antioxidant factors in pancreatic ductal adenocarcinoma (PDAC)- and hepatocellular carcinoma (HCC)-derived cells. In addition, ZZW-115-treatment increases the accumulation of hydroperoxided lipids in these cells. We also found that a loss of activity and strong deregulation of key enzymes involved in the GSH- and GPX-dependent antioxidant systems upon ZZW-115 treatment. These results have been validated in xenografts induced with PDAC- and HCC-derived cells in nude mice during the treatment with ZZW-115. More importantly, we demonstrate that ZZW-115-induced mitochondrial morphological changes, compatible with the ferroptotic process, as well as mitochondrial network disorganization and strong mitochondrial metabolic dysfunction, which are rescued by both Fer-1 and N-acetylcysteine (NAC). Of note, the expression of TFAM, a key regulator of mitochondrial biogenesis, is downregulated by ZZW-115. Forced expression of TFAM is able to rescue morphological and functional mitochondrial alterations, ROS production, and cell death induced by ZZW-115 or genetic inhibition of NUPR1. Altogether, these results demonstrate that the mitochondrial cell death mediated by NUPR1 inhibitor ZZW-115 is fully rescued by Fer-1 but also via TFAM complementation. In conclusion, TFAM could be considered as an antagonist of the ferroptotic cell death.
Collapse
|
265
|
Liu X, Zhang Y, Ren Y, Li J. Melatonin prevents allergic airway inflammation in epicutaneously sensitized mice. Biosci Rep 2021; 41:BSR20210398. [PMID: 34522948 PMCID: PMC8458693 DOI: 10.1042/bsr20210398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 11/17/2022] Open
Abstract
PURPOSE The pathological process of atopic dermatitis (AD) progressing into other types of allergic diseases such as asthma and allergic rhinitis during the first several years of life is often referred to as the atopic march. Although the phenomenon of atopic march has been recognized for decades, how asthma stems from AD is still not fully understood, confounding a universal strategy to effectively protect people from the atopic march. METHODS We established experimental atopic march mice by first inducing allergic dermatitis with 0.5% fluorescein isothiocyante (FITC) applied to the skin, followed by an ovalbumin (OVA) airway challenge. In addition, by examining serum immunoglobulin (Ig) concentrations, airway cytokines, the levels of oxidative stress markers, histopathological changes in lung tissue and airway hyperresponsiveness (AHR), we were able to validate the successful establishment of the model. Furthermore, by detecting the attenuating effects of melatonin (MT) and the levels of oxidative stress in the atopic march mice, we explored the potential molecular mechanisms involved in the development of atopic march. RESULTS By successfully establishing an experimental atopic march mouse model, we were able to demonstrate that overproduction of oxidative stress in the lung significantly up-regulated the activation of nuclear factor-κB (NF-κB) signaling pathways causing thymic stromal lymphopoietin (TSLP) release, which further promotes the development of atopic march. CONCLUSIONS To mitigate the development of the atopic march, antioxidants such as MT may be imperative to inhibit NF-κB activation in the lung, especially after the onset of AD.
Collapse
Affiliation(s)
- Xudong Liu
- Department of Food Science and Engineering, Moutai Institute, Renhuai 564507, China
| | - Yuchao Zhang
- Department of Food Science and Engineering, Moutai Institute, Renhuai 564507, China
| | - Yaolin Ren
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Jinquan Li
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| |
Collapse
|
266
|
Abstract
Abstract
Ebselen is a well-known synthetic compound mimicking glutathione peroxidase (GPx), which catalyses some vital reactions that protect against oxidative damage. Based on a large number of in vivo and in vitro studies, various mechanisms have been proposed to explain its actions on multiple targets. It targets thiol-related compounds, including cysteine, glutathione, and thiol proteins (e.g., thioredoxin and thioredoxin reductase). Owing to this, ebselen is a unique multifunctional agent with important effects on inflammation, apoptosis, oxidative stress, cell differentiation, immune regulation and neurodegenerative disease, with anti-microbial, detoxifying and anti-tumour activity. This review summarises the current understanding of the multiple biological processes and molecules targeted by ebselen, and its pharmacological applications.
Collapse
|
267
|
Lai Y, Zhang T, Song W, Li Z, Lin W. Evaluation of Cell Viability with a Single Fluorescent Probe Based on Two Kinds of Fluorescence Signal Modes. Anal Chem 2021; 93:12487-12493. [PMID: 34455772 DOI: 10.1021/acs.analchem.1c02911] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Accurate evaluation of cell viability is important for dosage tests of anticancer drugs, pathology, and numerous biological experiments. However, due to the serious insufficieny of fluorescent probes, which can distinguish various cell states, the study of cell viability is immensely limited. To resolve this issue, we design and synthesize a new probe ACD-E to monitor cell viability with two kinds of fluorescence signal modes, the first single fluorescent probe that can distinguish three different cell states and furnish accurate information in biological experiments. ACD-E can discriminate live and dead cells in a dual-color mode by evaluating cell mitochondrial esterase activity and can also discriminate live and early necrosis cells by determining mitochondrial viscosity in a "turn-on" mode in the near-infrared region. Significantly, the novel ACD-E can also distinguish cell viability in vivo. This work establishes a robust strategy for monitoring multiple cell states using a single fluorescent probe.
Collapse
Affiliation(s)
- Youbo Lai
- Guangxi Key Laboratory of Electrochemical Energy Materials, Institute of Optical Materials and Chemical Biology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P. R. China
| | - Tengteng Zhang
- Guangxi Key Laboratory of Electrochemical Energy Materials, Institute of Optical Materials and Chemical Biology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P. R. China
| | - Wenhui Song
- Guangxi Key Laboratory of Electrochemical Energy Materials, Institute of Optical Materials and Chemical Biology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P. R. China
| | - Zihong Li
- Guangxi Key Laboratory of Electrochemical Energy Materials, Institute of Optical Materials and Chemical Biology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P. R. China
| | - Weiying Lin
- Guangxi Key Laboratory of Electrochemical Energy Materials, Institute of Optical Materials and Chemical Biology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P. R. China
| |
Collapse
|
268
|
Farooq A, Richman CM, Swain SM, Shahid RA, Vigna SR, Liddle RA. The Role of Phosphate in Alcohol-Induced Experimental Pancreatitis. Gastroenterology 2021; 161:982-995.e2. [PMID: 34051238 PMCID: PMC8380702 DOI: 10.1053/j.gastro.2021.05.048] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Heavy alcohol consumption is a common cause of acute pancreatitis; however, alcohol abuse does not always result in clinical pancreatitis. As a consequence, the factors responsible for alcohol-induced pancreatitis are not well understood. In experimental animals, it has been difficult to produce pancreatitis with alcohol. Clinically, alcohol use predisposes to hypophosphatemia, and hypophosphatemia has been observed in some patients with acute pancreatitis. Because of abundant protein synthesis, the pancreas has high metabolic demands, and reduced mitochondrial function leads to organelle dysfunction and pancreatitis. We proposed, therefore, that phosphate deficiency might limit adenosine triphosphate synthesis and thereby contribute to alcohol-induced pancreatitis. METHODS Mice were fed a low-phosphate diet (LPD) before orogastric administration of ethanol. Direct effects of phosphate and ethanol were evaluated in vitro in isolated mouse pancreatic acini. RESULTS LPD reduced serum phosphate levels. Intragastric administration of ethanol to animals maintained on an LPD caused severe pancreatitis that was ameliorated by phosphate repletion. In pancreatic acinar cells, low-phosphate conditions increased susceptibility to ethanol-induced cellular dysfunction through decreased bioenergetic stores, specifically affecting total cellular adenosine triphosphate and mitochondrial function. Phosphate supplementation prevented ethanol-associated cellular injury. CONCLUSIONS Phosphate status plays a critical role in predisposition to and protection from alcohol-induced acinar cell dysfunction and the development of acute alcohol-induced pancreatitis. This finding may explain why pancreatitis develops in only some individuals with heavy alcohol use and suggests a potential novel therapeutic approach to pancreatitis. Finally, an LPD plus ethanol provides a new model for studying alcohol-associated pancreatic injury.
Collapse
Affiliation(s)
- Ahmad Farooq
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Courtney M Richman
- School of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Sandip M Swain
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Rafiq A Shahid
- Department of Pathology, Brown University, Providence, Rhode Island
| | - Steven R Vigna
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Rodger A Liddle
- Department of Medicine, Duke University Medical Center, Durham, North Carolina; Department of Veterans Affairs Health Care System, Durham, North Carolina.
| |
Collapse
|
269
|
MacDonald IJ, Tsai HC, Chang AC, Huang CC, Yang SF, Tang CH. Melatonin Inhibits Osteoclastogenesis and Osteolytic Bone Metastasis: Implications for Osteoporosis. Int J Mol Sci 2021; 22:ijms22179435. [PMID: 34502344 PMCID: PMC8430520 DOI: 10.3390/ijms22179435] [Citation(s) in RCA: 16] [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: 07/28/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/16/2022] Open
Abstract
Osteoblasts and osteoclasts are major cellular components in the bone microenvironment and they play a key role in the bone turnover cycle. Many risk factors interfere with this cycle and contribute to bone-wasting diseases that progressively destroy bone and markedly reduce quality of life. Melatonin (N-acetyl-5-methoxy-tryptamine) has demonstrated intriguing therapeutic potential in the bone microenvironment, with reported effects that include the regulation of bone metabolism, acceleration of osteoblastogenesis, inhibition of osteoclastogenesis and the induction of apoptosis in mature osteoclasts, as well as the suppression of osteolytic bone metastasis. This review aims to shed light on molecular and clinical evidence that points to possibilities of melatonin for the treatment of both osteoporosis and osteolytic bone metastasis. It appears that the therapeutic qualities of melatonin supplementation may enable existing antiresorptive osteoporotic drugs to treat osteolytic metastasis.
Collapse
Affiliation(s)
- Iona J. MacDonald
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan; (I.J.M.); (H.-C.T.)
| | - Hsiao-Chi Tsai
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan; (I.J.M.); (H.-C.T.)
| | - An-Chen Chang
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei City 111, Taiwan;
| | - Chien-Chung Huang
- School of Medicine, China Medical University, Taichung 40402, Taiwan;
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung 40447, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan; (I.J.M.); (H.-C.T.)
- School of Medicine, China Medical University, Taichung 40402, Taiwan;
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung 41354, Taiwan
- Correspondence: ; Tel.: +886-2205-2121 (ext. 7726)
| |
Collapse
|
270
|
NMDA receptors elicit flux-independent intracellular Ca 2+ signals via metabotropic glutamate receptors and flux-dependent nitric oxide release in human brain microvascular endothelial cells. Cell Calcium 2021; 99:102454. [PMID: 34454368 DOI: 10.1016/j.ceca.2021.102454] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/28/2021] [Accepted: 08/12/2021] [Indexed: 12/20/2022]
Abstract
The excitatory neurotransmitter glutamate gates post-synaptic N-methyl-d-aspartate (NMDA) receptors (NMDARs) to mediate extracellular Ca2+ entry and stimulate neuronal nitric oxide (NO) synthase to release NO and trigger neurovascular coupling (NVC). Neuronal and glial NMDARs may also operate in a flux-independent manner, although it is unclear whether their non-ionotropic mode of action is involved in NVC. Recently, endothelial NMDARs were found to trigger Ca2+-dependent NO production and induce NVC, but the underlying mode of signaling remains elusive. Herein, we report that GluN1 protein, as well as GluN2C and GluN3B transcripts and proteins, were expressed and that NMDA did not elicit inward currents, but induced a dose-dependent increase in intracellular Ca2+ concentration ([Ca2+]i) in the human brain microvascular endothelial cell line, hCMEC/D3. A multidisciplinary approach, including live cell imaging, whole-cell patch-clamp recordings, pharmacological manipulation and gene targeting, revealed that NMDARs increase the [Ca2+]i in a flux-independent manner in hCMEC/D3 cells. The Ca2+ response to NMDA was triggered by endogenous Ca2+ release from the endoplasmic reticulum and the lysosomal Ca2+ stores and sustained by store-operated Ca2+ entry. Unexpectedly, pharmacological and genetic blockade of mGluR1 and mGluR5 dramatically impaired NMDARs-mediated Ca2+ signals. These findings indicate that NMDARs may increase the endothelial [Ca2+]i in a flux-independent manner via group 1 mGluRs. However, imaging of DAF-FM fluorescence revealed that NMDARs may also induce Ca2+-dependent NO release by signaling in a flux-dependent manner. These findings, therefore, shed novel light on the mechanisms whereby brain microvascular endothelium decodes glutamatergic signaling and regulates NVC.
Collapse
|
271
|
Zhang J, Lu X, Liu M, Fan H, Zheng H, Zhang S, Rahman N, Wołczyński S, Kretowski A, Li X. Melatonin inhibits inflammasome-associated activation of endothelium and macrophages attenuating pulmonary arterial hypertension. Cardiovasc Res 2021; 116:2156-2169. [PMID: 31774487 DOI: 10.1093/cvr/cvz312] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/25/2019] [Accepted: 11/25/2019] [Indexed: 01/08/2023] Open
Abstract
AIMS Pulmonary arterial hypertension (PAH) is a pathophysiological syndrome associated with pulmonary/systemic inflammation. Melatonin relieves PAH, but the molecular mode of action remains unclear. Here, we investigated the role of melatonin in normalizing vascular homeostasis. METHODS AND RESULTS Light-time mean serum melatonin concentration was lower in patients with PAH than in normal controls [11.06 ± 3.44 (7.13-15.6) vs. 14.55 ± 1.28 (8.0-19.4) pg/mL], which was negatively correlated with increased serum levels of interleukin-1β (IL-1β) in patients with PAH. We showed that inflammasomes were activated in the PAH mice model and that melatonin attenuated IL-1β secretion. On one hand, melatonin reduced the number of macrophages in lung by inhibiting the endothelial chemokines and adhesion factors. Moreover, use of Il1r-/- mice, Caspase1/11-/- mice, and melatonin-treated mice revealed that melatonin reduced hypoxia-induced vascular endothelial leakage in the lung. On the other hand, we verified that melatonin reduced the formation of inflammasome multiprotein complexes by modulating calcium ions in macrophages using a live cell station, and melatonin decreased inositol triphosphate and increased cAMP. Furthermore, knockdown of melatonin membrane receptors blocked melatonin function, and a melatonin membrane receptors agonist inactivated inflammasomes in macrophages. CONCLUSION Melatonin attenuated inflammasome-associated vascular disorders by directly improving endothelial leakage and decreasing the formation of inflammasome multiprotein complexes in macrophages. Taken together, our data provide a theoretical basis for applying melatonin clinically, and inflammasomes may be a possible target of PAH treatment.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xiaohui Lu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Mei Liu
- Department of Pathology, Chinese PLA General Hospital, Beijing 102628, China
| | - Hanlu Fan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Han Zheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Shanshan Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Nafis Rahman
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Sławomir Wołczyński
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Adam Kretowski
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Białystok, Poland
| | - Xiangdong Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.,Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
272
|
Joshi A, Upadhyay KK, Vohra A, Shirsath K, Devkar R. Melatonin induces Nrf2-HO-1 reprogramming and corrections in hepatic core clock oscillations in Non-alcoholic fatty liver disease. FASEB J 2021; 35:e21803. [PMID: 34365685 DOI: 10.1096/fj.202002556rrr] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/20/2022]
Abstract
Melatonin pleiotropically regulates physiological events and has a putative regulatory role in the circadian clock desynchrony-mediated Non-alcoholic fatty liver disease (NAFLD). In this study, we investigated perturbations in the hepatic circadian clock gene, and Nrf2-HO-1 oscillations in conditions of high-fat high fructose (HFHF) diet and/or jet lag (JL)-mediated NAFLD. Melatonin treatment (100 µM) to HepG2 cells led to an improvement in oscillatory pattern of clock genes (Clock, Bmal1, and Per) in oleic acid (OA)-induced circadian desynchrony, while Cry, Nrf2, and HO-1 remain oblivious of melatonin treatment that was also validated by circwave analysis. C57BL/6J mice subjected to HFHF and/or JL, and treated with melatonin showed an improvement in the profile of lipid regulatory genes (CPT-1, PPARa, and SREBP-1c), liver function (AST and ALT) and histomorphology of fatty liver. A detailed scrutiny revealed that hepatic mRNA and protein profiles of Bmal1 (at ZT6) and Clock (at ZT12) underwent corrective changes in oscillations, but moderate corrections were recorded in other components of clock genes (Per1, Per2, and Cry2). Melatonin induced changes in oscillations of anti-oxidant genes (Nrf2, HO-1, and Keap1) subtly contributed in the overall improvement in NAFLD recorded herein. Taken together, melatonin induced reprograming of hepatic core clock and Nrf2-HO-1 genes leads to an improvement in HFHF/JL-induced NAFLD.
Collapse
Affiliation(s)
- Apeksha Joshi
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Kapil K Upadhyay
- Department of Internal medicine, Division of Gastroenterology and Hepatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Aliasgar Vohra
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Kavita Shirsath
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Ranjitsinh Devkar
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
273
|
Wang B, Shan X, Lv S, Zha L, Zhang C, Dong Q, Chen W. Preparation, Characterization, and In Vitro/In Vivo Evaluation of 3-O-β-D-Galactosylated Resveratrol-Loaded Polydopamine Nanoparticles. AAPS PharmSciTech 2021; 22:220. [PMID: 34405290 DOI: 10.1208/s12249-021-02079-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/23/2021] [Indexed: 11/30/2022] Open
Abstract
3-O-β-D-galactosylated resveratrol (Gal-Res) was synthesized from resveratrol (Res) and 3-O-β-D-galactose (Gal) in our previous study. In order to improve the pH sensitivity and bioavailability of Gal-Res, Gal-Res nanoparticles (Gal-Res NPs) were prepared using polydopamine (PDA) as a drug carrier. The drug loading (DL %) and entrapment efficiency (EE %) of Gal-Res NPs were 46.80% and 88.06%. The average particle size, polydispersity index (PDI), and Zeta potential of Gal-Res NPs were 179.38 ± 2.83 nm, 0.129 ± 0.013, and - 28.05 ± 0.36 mV, respectively. The transmission electron microscope (TEM) showed that Gal-Res NPs had uniform spherical morphology. Compared with the fast release of raw Gal-Res, the in vitro release of Gal-Res NPs was slow and pH-sensitive. The results of the blood vessel irritation and hemolysis test demonstrated that Gal-Res NPs had good hemocompatibility. The pharmacokinetics study in rats showed that area under the curve of plasma drug concentration time (AUC0→600) and half-life (t1/2) of Gal-Res NPs were enhanced 1.82-fold and 2.19-fold higher than those of raw Gal-Res. The in vivo biodistribution results showed that Gal-Res NPs were more distributed in liver tissue than Gal-Res. Gal-Res NPs with high bioavailability and liver accumulation were hopeful drug delivery systems (DDS) to treat liver diseases.
Collapse
|
274
|
Melatonin Promotes In Vitro Maturation of Vitrified-Warmed Mouse Germinal Vesicle Oocytes, Potentially by Reducing Oxidative Stress through the Nrf2 Pathway. Animals (Basel) 2021; 11:ani11082324. [PMID: 34438783 PMCID: PMC8388487 DOI: 10.3390/ani11082324] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/30/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Cryopreservation of oocytes can cause high oxidative stress, reduce the quality of vitrified-warmed oocytes, and seriously hinder the application of oocyte cryopreservation technology in production and medicine. In this work, we found for the first time that melatonin can exert antioxidant effects through receptors and regulate the Nrf2 antioxidant pathway to respond to oxidative stress of vitrified-warmed oocytes, thereby improving both oocyte quality and the potential for subsequent development. The results illustrated the molecular mechanism of melatonin’s antioxidant effect in vitrified-warmed oocytes and provided a theoretical basis for the application of melatonin in the cryopreservation of oocytes. These findings are of great significance for the further application of oocyte cryopreservation technology to production and assisted reproduction in the future. Abstract Previously it was reported that melatonin could mitigate oxidative stress caused by oocyte cryopreservation; however, the underlying molecular mechanisms which cause this remain unclear. The objective was to explore whether melatonin could reduce oxidative stress during in vitro maturation of vitrified-warmed mouse germinal vesicle (GV) oocytes through the Nrf2 signaling pathway or its receptors. During in vitro maturation of vitrified-warmed mouse GV oocytes, there were decreases (p < 0.05) in the development rates of metaphase I (MI) oocytes and metaphase II (MII) and spindle morphology grades; increases (p < 0.05) in the reactive oxygen species (ROS) levels; and decreases (p < 0.05) in expressions of Nrf2 signaling pathway-related genes (Nrf2, SOD1) and proteins (Nrf2, HO-1). However, adding 10−7 mol/L melatonin to both the warming solution and maturation solutions improved (p < 0.05) these indicators. When the Nrf2 protein was specifically inhibited by Brusatol, melatonin did not increase development rates, spindle morphology grades, genes, or protein expressions, nor did it reduce vitrification-induced intracellular oxidative stress in GV oocytes during in vitro maturation. In addition, when melatonin receptors were inhibited by luzindole, the ability of melatonin to scavenge intracellular ROS was decreased, and the expressions of genes (Nrf2, SOD1) and proteins (Nrf2, HO-1) were not restored to control levels. Therefore, we concluded that 10−7 mol/L melatonin acted on the Nrf2 signaling pathway through its receptors to regulate the expression of genes (Nrf2, SOD1) and proteins (Nrf2, HO-1), and mitigate intracellular oxidative stress, thereby enhancing in vitro development of vitrified-warmed mouse GV oocytes.
Collapse
|
275
|
Satpathy S, Krug K, Jean Beltran PM, Savage SR, Petralia F, Kumar-Sinha C, Dou Y, Reva B, Kane MH, Avanessian SC, Vasaikar SV, Krek A, Lei JT, Jaehnig EJ, Omelchenko T, Geffen Y, Bergstrom EJ, Stathias V, Christianson KE, Heiman DI, Cieslik MP, Cao S, Song X, Ji J, Liu W, Li K, Wen B, Li Y, Gümüş ZH, Selvan ME, Soundararajan R, Visal TH, Raso MG, Parra ER, Babur Ö, Vats P, Anand S, Schraink T, Cornwell M, Rodrigues FM, Zhu H, Mo CK, Zhang Y, da Veiga Leprevost F, Huang C, Chinnaiyan AM, Wyczalkowski MA, Omenn GS, Newton CJ, Schurer S, Ruggles KV, Fenyö D, Jewell SD, Thiagarajan M, Mesri M, Rodriguez H, Mani SA, Udeshi ND, Getz G, Suh J, Li QK, Hostetter G, Paik PK, Dhanasekaran SM, Govindan R, Ding L, Robles AI, Clauser KR, Nesvizhskii AI, Wang P, Carr SA, Zhang B, Mani DR, Gillette MA. A proteogenomic portrait of lung squamous cell carcinoma. Cell 2021; 184:4348-4371.e40. [PMID: 34358469 PMCID: PMC8475722 DOI: 10.1016/j.cell.2021.07.016] [Citation(s) in RCA: 220] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 04/26/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
Lung squamous cell carcinoma (LSCC) remains a leading cause of cancer death with few therapeutic options. We characterized the proteogenomic landscape of LSCC, providing a deeper exposition of LSCC biology with potential therapeutic implications. We identify NSD3 as an alternative driver in FGFR1-amplified tumors and low-p63 tumors overexpressing the therapeutic target survivin. SOX2 is considered undruggable, but our analyses provide rationale for exploring chromatin modifiers such as LSD1 and EZH2 to target SOX2-overexpressing tumors. Our data support complex regulation of metabolic pathways by crosstalk between post-translational modifications including ubiquitylation. Numerous immune-related proteogenomic observations suggest directions for further investigation. Proteogenomic dissection of CDKN2A mutations argue for more nuanced assessment of RB1 protein expression and phosphorylation before declaring CDK4/6 inhibition unsuccessful. Finally, triangulation between LSCC, LUAD, and HNSCC identified both unique and common therapeutic vulnerabilities. These observations and proteogenomics data resources may guide research into the biology and treatment of LSCC.
Collapse
Affiliation(s)
- Shankha Satpathy
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA.
| | - Karsten Krug
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Pierre M Jean Beltran
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Sara R Savage
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Yongchao Dou
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Boris Reva
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - M Harry Kane
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Shayan C Avanessian
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Suhas V Vasaikar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Azra Krek
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jonathan T Lei
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eric J Jaehnig
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Yifat Geffen
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Erik J Bergstrom
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Vasileios Stathias
- Sylvester Comprehensive Cancer Center and Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Karen E Christianson
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - David I Heiman
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Marcin P Cieslik
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Song Cao
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Xiaoyu Song
- Department of Population Health Science and Policy, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jiayi Ji
- Department of Population Health Science and Policy, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Wenke Liu
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Kai Li
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bo Wen
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yize Li
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tanvi H Visal
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maria G Raso
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Edwin Roger Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Özgün Babur
- Computer Science Department, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Pankaj Vats
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shankara Anand
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Tobias Schraink
- Institute for Systems Genetics and Department of Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - MacIntosh Cornwell
- Institute for Systems Genetics and Department of Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | | | - Houxiang Zhu
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Chia-Kuei Mo
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Yuping Zhang
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Chen Huang
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Gilbert S Omenn
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Stephan Schurer
- Sylvester Comprehensive Cancer Center and Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Kelly V Ruggles
- Institute for Systems Genetics and Department of Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - David Fenyö
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Scott D Jewell
- Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Mathangi Thiagarajan
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Namrata D Udeshi
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Gad Getz
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - James Suh
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Qing Kay Li
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Medical Institutions, Baltimore, MD 21224, USA
| | | | - Paul K Paik
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Ramaswamy Govindan
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Li Ding
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Karl R Clauser
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Steven A Carr
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA.
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| | - D R Mani
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA.
| | - Michael A Gillette
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02115, USA.
| |
Collapse
|
276
|
Bonvento G, Bolaños JP. Astrocyte-neuron metabolic cooperation shapes brain activity. Cell Metab 2021; 33:1546-1564. [PMID: 34348099 DOI: 10.1016/j.cmet.2021.07.006] [Citation(s) in RCA: 250] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/11/2021] [Accepted: 07/03/2021] [Indexed: 12/12/2022]
Abstract
The brain has almost no energy reserve, but its activity coordinates organismal function, a burden that requires precise coupling between neurotransmission and energy metabolism. Deciphering how the brain accomplishes this complex task is crucial to understand central facets of human physiology and disease mechanisms. Each type of neural cell displays a peculiar metabolic signature, forcing the intercellular exchange of metabolites that serve as both energy precursors and paracrine signals. The paradigm of this biological feature is the astrocyte-neuron couple, in which the glycolytic metabolism of astrocytes contrasts with the mitochondrial oxidative activity of neurons. Astrocytes generate abundant mitochondrial reactive oxygen species and shuttle to neurons glycolytically derived metabolites, such as L-lactate and L-serine, which sustain energy needs, conserve redox status, and modulate neurotransmitter-receptor activity. Conversely, early disruption of this metabolic cooperation may contribute to the initiation or progression of several neurological diseases, thus requiring innovative therapies to preserve brain energetics.
Collapse
Affiliation(s)
- Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France.
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Institute of Biomedical Research of Salamanca, Salamanca, Spain
| |
Collapse
|
277
|
Haddad M, Hervé V, Ben Khedher MR, Rabanel JM, Ramassamy C. Glutathione: An Old and Small Molecule with Great Functions and New Applications in the Brain and in Alzheimer's Disease. Antioxid Redox Signal 2021; 35:270-292. [PMID: 33637005 DOI: 10.1089/ars.2020.8129] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Significance: Glutathione (GSH) represents the most abundant and the main antioxidant in the body with important functions in the brain related to Alzheimer's disease (AD). Recent Advances: Oxidative stress is one of the central mechanisms in AD. We and others have demonstrated the alteration of GSH levels in the AD brain, its important role in the detoxification of advanced glycation end-products and of acrolein, a by-product of lipid peroxidation. Recent in vivo studies found a decrease of GSH in several areas of the brain from control, mild cognitive impairment, and AD subjects, which are correlated with cognitive decline. Critical Issues: Several strategies were developed to restore its intracellular level with the l-cysteine prodrugs or the oral administration of γ-glutamylcysteine to prevent alterations observed in AD. To date, no benefit on GSH level or on oxidative biomarkers has been reported in clinical trials. Thus, it remains uncertain if GSH could be considered a potential preventive or therapeutic approach or a biomarker for AD. Future Directions: We address how GSH-coupled nanocarriers represent a promising approach for the functionalization of nanocarriers to overcome the blood/brain barrier (BBB) for the brain delivery of GSH while avoiding cellular toxicity. It is also important to address the presence of GSH in exosomes for its potential intercellular transfer or its shuttle across the BBB under certain conditions. Antioxid. Redox Signal. 35, 270-292.
Collapse
Affiliation(s)
- Mohamed Haddad
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada.,Institute on Nutrition and Functional Foods, Université Laval, Québec, Canada
| | - Vincent Hervé
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada.,Institute on Nutrition and Functional Foods, Université Laval, Québec, Canada
| | - Mohamed Raâfet Ben Khedher
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada.,Institute on Nutrition and Functional Foods, Université Laval, Québec, Canada
| | | | - Charles Ramassamy
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada.,Institute on Nutrition and Functional Foods, Université Laval, Québec, Canada
| |
Collapse
|
278
|
Du Z, Song Y, Chen X, Zhang W, Zhang G, Li H, Chang L, Wu Y. Knockdown of astrocytic Grin2a aggravates β-amyloid-induced memory and cognitive deficits through regulating nerve growth factor. Aging Cell 2021; 20:e13437. [PMID: 34291567 PMCID: PMC8373273 DOI: 10.1111/acel.13437] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/14/2021] [Accepted: 07/03/2021] [Indexed: 12/14/2022] Open
Abstract
Synapse degeneration correlates strongly with cognitive impairments in Alzheimer's disease (AD) patients. Soluble Amyloid-beta (Aβ) oligomers are thought as the major trigger of synaptic malfunctions. Our earlier studies have demonstrated that Aβ oligomers interfere with synaptic function through N-methyl-D-aspartate receptors (NMDARs). Our recent in vitro study found the neuroprotective role of astrocytic GluN2A in the promotion of synapse survival and identified nerve growth factor (NGF) derived from astrocytes, as a likely mediator of astrocytic GluN2A buffering against Aβ synaptotoxicity. Our present in vivo study focused on exploring the precise mechanism of astrocytic GluN2A influencing Aβ synaptotoxicity through regulating NGF. We generated an adeno-associated virus (AAV) expressing an astrocytic promoter (GfaABC1D) shRNA targeted to Grin2a (the gene encoding GluN2A) to perform astrocyte-specific Grin2a knockdown in the hippocampal dentate gyrus, after 3 weeks of virus vector expression, Aβ were bilaterally injected into the intracerebral ventricle. Our results showed that astrocyte-specific knockdown of Grin2a and Aβ application both significantly impaired spatial memory and cognition, which associated with the reduced synaptic proteins PSD95, synaptophysin and compensatory increased NGF. The reduced astrocytic GluN2A can counteract Aβ-induced compensatory protective increase of NGF through regulating pNF-κB, Furin and VAMP3, which modulating the synthesis, mature and secretion of NGF respectively. Our present data reveal, for the first time, a novel mechanism of astrocytic GluN2A in exerting protective effects on synapses at the early stage of Aβ exposure, which may contribute to establish new targets for AD prevention and early therapy.
Collapse
Affiliation(s)
- Zunshu Du
- Beijing Key Laboratory of Neural Regeneration and Repair Department of Anatomy School of Basic Medical Sciences Beijing Institute of Brain Disorders Capital Medical University Beijing China
| | - Yizhi Song
- Beijing Key Laboratory of Neural Regeneration and Repair Department of Anatomy School of Basic Medical Sciences Beijing Institute of Brain Disorders Capital Medical University Beijing China
| | - Xinyue Chen
- Beijing Key Laboratory of Neural Regeneration and Repair Department of Anatomy School of Basic Medical Sciences Beijing Institute of Brain Disorders Capital Medical University Beijing China
| | - Wanning Zhang
- Beijing Key Laboratory of Neural Regeneration and Repair Department of Anatomy School of Basic Medical Sciences Beijing Institute of Brain Disorders Capital Medical University Beijing China
| | - Guitao Zhang
- Beijing Key Laboratory of Neural Regeneration and Repair Department of Anatomy School of Basic Medical Sciences Beijing Institute of Brain Disorders Capital Medical University Beijing China
| | - Hui Li
- Beijing Key Laboratory of Neural Regeneration and Repair Department of Anatomy School of Basic Medical Sciences Beijing Institute of Brain Disorders Capital Medical University Beijing China
| | - Lirong Chang
- Beijing Key Laboratory of Neural Regeneration and Repair Department of Anatomy School of Basic Medical Sciences Beijing Institute of Brain Disorders Capital Medical University Beijing China
| | - Yan Wu
- Beijing Key Laboratory of Neural Regeneration and Repair Department of Anatomy School of Basic Medical Sciences Beijing Institute of Brain Disorders Capital Medical University Beijing China
| |
Collapse
|
279
|
Manko BO, Bilonoha OO, Voloshyn DM, Zub AM, Ivasechko II, Manko VV. Pyruvate and Glutamine Define the Effects of Cholecystokinin and Ethanol on Mitochondrial Oxidation, Necrosis, and Morphology of Rat Pancreatic Acini. Pancreas 2021; 50:972-981. [PMID: 34629447 DOI: 10.1097/mpa.0000000000001864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The objective of this study was to test whether pyruvate and glutamine affect the ethanol and cholecystokinin (CCK) effects on the mitochondrial function, viability, and morphology of rat pancreatic acini. METHODS Respiration was measured with Clark oxygen electrode. Mitochondrial membrane potential, reduced nicotinamide adenine dinucleotide (phosphate) (NAD(P)H), cell morphology, and viability were studied with fluorescence microscopy. RESULTS In vitro, CCK (0.1 nM) caused pyruvate-dependent stimulation of basal and uncoupled respiration, and the effects were abolished by ethanol (20 mM). The combination of ethanol with CCK (2 hours) caused necrosis of approximately 40% acinar cells in medium with glucose, but not with pyruvate and/or glutamine. Cholecystokinin (10 nM) or ethanol with 0.1 nM CCK caused plasma membrane blebbing not related to apoptosis only when both glutamine and pyruvate were present. Glutamine, but not pyruvate, decreased NAD(P)H level and prevented the effects of ethanol with CCK on mitochondrial membrane potential and NAD(P)H, but, in combination with CCK and ethanol, decreased the uncoupled respiration. In vivo, the combination of ethanol (4 g/kg) and CCK (20 pmol/kg) suppressed basal and uncoupled respiration and caused acinar cell blebbing, but not necrosis. CONCLUSIONS The lack of sufficient substrate supply in vitro makes pancreatic acinar cells susceptible to necrosis caused by ethanol and CCK in clinically relevant concentrations.
Collapse
Affiliation(s)
- Bohdan O Manko
- From the Human and Animal Physiology Department, Ivan Franko National University of Lviv
| | - Olha O Bilonoha
- From the Human and Animal Physiology Department, Ivan Franko National University of Lviv
| | - Dariia M Voloshyn
- From the Human and Animal Physiology Department, Ivan Franko National University of Lviv
| | - Anastasiia M Zub
- From the Human and Animal Physiology Department, Ivan Franko National University of Lviv
| | - Iryna I Ivasechko
- Institute of Cell Biology of National Academy of Sciences of Ukraine, Lviv, Ukraine
| | - Volodymyr V Manko
- From the Human and Animal Physiology Department, Ivan Franko National University of Lviv
| |
Collapse
|
280
|
Huang K, Wang C, Vagts C, Raguveer V, Finn PW, Perkins DL. Long non-coding RNAs (lncRNAs) NEAT1 and MALAT1 are differentially expressed in severe COVID-19 patients: An integrated single cell analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.03.26.21254445. [PMID: 33821282 PMCID: PMC8020982 DOI: 10.1101/2021.03.26.21254445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hyperactive and damaging inflammation is a hallmark of severe rather than mild COVID-19 syndrome. To uncover key inflammatory differentiators between severe and mild COVID-19 disease, we applied an unbiased single-cell transcriptomic analysis. We integrated a bronchoalveolar lavage (BAL) dataset with a peripheral blood mononuclear cell dataset (PBMC) and analyzed the combined cell population, focusing on genes associated with disease severity. Distinct cell populations were detected in both BAL and PBMC where the immunomodulatory long non-coding RNAs (lncRNAs) NEAT1 and MALAT1 were highly differentially expressed between mild and severe patients. The detection of other severity associated genes involved in cellular stress response and apoptosis regulation suggests that the pro-inflammatory functions of these lncRNAs may foster cell stress and damage. The lncRNAs NEAT1 and MALAT1 are potential components of immune dysregulation in COVID-19 that may provide targets for severity related diagnostic measures or therapy.
Collapse
Affiliation(s)
- Kai Huang
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago IL, 60612 United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago IL, 60612 United States
| | - Catherine Wang
- College of Medicine, University of Illinois at Chicago, Chicago IL, 60612 United States
| | - Christen Vagts
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago IL, 60612 United States
| | - Vanitha Raguveer
- College of Medicine, University of Illinois at Chicago, Chicago IL, 60612 United States
| | - Patricia W. Finn
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago IL, 60612 United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago IL, 60612 United States
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago IL, 60612 United States
| | - David L. Perkins
- Department of Bioengineering, University of Illinois at Chicago, Chicago IL, 60612 United States
- Division of Nephrology, Department of Medicine, University of Illinois at Chicago, Chicago IL, 60612 United States
- Department of Surgery, University of Illinois at Chicago, Chicago IL, 60612 United States
| |
Collapse
|
281
|
Estaras M, Gonzalez A. Modulation of cell physiology under hypoxia in pancreatic cancer. World J Gastroenterol 2021; 27:4582-4602. [PMID: 34366624 PMCID: PMC8326256 DOI: 10.3748/wjg.v27.i28.4582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/28/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
In solid tumors, the development of vasculature is, to some extent, slower than the proliferation of the different types of cells that form the tissue, both cancer and stroma cells. As a consequence, the oxygen availability is compromised and the tissue evolves toward a condition of hypoxia. The presence of hypoxia is variable depending on where the cells are localized, being less extreme at the periphery of the tumor and more severe in areas located deep within the tumor mass. Surprisingly, the cells do not die. Intracellular pathways that are critical for cell fate such as endoplasmic reticulum stress, apoptosis, autophagy, and others are all involved in cellular responses to the low oxygen availability and are orchestrated by hypoxia-inducible factor. Oxidative stress and inflammation are critical conditions that develop under hypoxia. Together with changes in cellular bioenergetics, all contribute to cell survival. Moreover, cell-to-cell interaction is established within the tumor such that cancer cells and the microenvironment maintain a bidirectional communication. Additionally, the release of extracellular vesicles, or exosomes, represents short and long loops that can convey important information regarding invasion and metastasis. As a result, the tumor grows and its malignancy increases. Currently, one of the most lethal tumors is pancreatic cancer. This paper reviews the most recent advances in the knowledge of how cells grow in a pancreatic tumor by adapting to hypoxia. Unmasking the physiological processes that help the tumor increase its size and their regulation will be of major relevance for the treatment of this deadly tumor.
Collapse
Affiliation(s)
- Matias Estaras
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres 10003, Spain
| | - Antonio Gonzalez
- Department of Physiology, Cell Biology and Communication Research Group, University of Extremadura, Caceres 10003, Spain
| |
Collapse
|
282
|
Qin DZ, Cai H, He C, Yang DH, Sun J, He WL, Li BL, Hua JL, Peng S. Melatonin relieves heat-induced spermatocyte apoptosis in mouse testes by inhibition of ATF6 and PERK signaling pathways. Zool Res 2021; 42:514-524. [PMID: 34254745 PMCID: PMC8317181 DOI: 10.24272/j.issn.2095-8137.2021.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Normal spermatogenic processes require the scrotal temperature to be lower than that of the body as excessive heat affects spermatogenesis in the testes, reduces sperm quality and quantity, and even causes infertility. Endoplasmic reticulum stress (ERS) is a crucial factor in many pathologies. Although several studies have linked ERS to heat stress, researchers have not yet determined which ERS signaling pathways contribute to heat-induced testicular damage. Melatonin activates antioxidant enzymes, scavenges free radicals, and protects the testes from inflammation; however, few studies have reported on the influence of melatonin on heat-induced testicular damage. Using a murine model of testicular hyperthermia, we observed that heat stress causes both ERS and apoptosis in the testes, especially in the spermatocytes. These observations were confirmed using the mouse spermatocyte cell line GC2, where the Atf6 and Perk signaling pathways were activated during heat stress. Knockout of the above genes effectively reduced spermatocyte damage caused by heat stress. Pretreatment with melatonin alleviated heat-induced apoptosis by inhibiting the Atf6 and Perk signaling pathways. This mitigation was dependent on the melatonin receptors. In vivo experiments verified that melatonin treatment relieved heat-induced testicular damage. In conclusion, our results demonstrated that ATF6 and PERK are important mediators for heat-induced apoptosis, which can be prevented by melatonin treatment. Thus, our study highlights melatonin as a potential therapeutic agent in mammals for subfertility/infertility induced by testicular hyperthermia.
Collapse
Affiliation(s)
- De-Zhe Qin
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Hui Cai
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Chen He
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Dong-Hui Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Jing Sun
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Wen-Lai He
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Ba-Lun Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Jin-Lian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Sha Peng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China. E-mail:
| |
Collapse
|
283
|
Jaworek AK, Szepietowski JC, Hałubiec P, Wojas-Pelc A, Jaworek J. Melatonin as an Antioxidant and Immunomodulator in Atopic Dermatitis-A New Look on an Old Story: A Review. Antioxidants (Basel) 2021; 10:antiox10081179. [PMID: 34439427 PMCID: PMC8388892 DOI: 10.3390/antiox10081179] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/16/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022] Open
Abstract
Atopic dermatitis (AD) is common inflammatory dermatosis, typically with chronic and recurrent course, which significantly reduces the quality of life. Sleep disturbances are considered to be remarkably burdensome ailments in patients with AD, and are routinely included during assessment of disease severity. Therefore, endogenous substances engaged in the control of circadian rhythms might be important in pathogenesis of AD and, possibly, be used as biomarkers of disease severity or even in development of novel therapies. Melatonin (MT), the indoleamine produced by pineal gland (but also by multiple other tissues, including skin), plays a pivotal role in maintaining the sleep/wake homeostasis. Additionally, it possesses strong antioxidant and anti-inflammatory properties, which might directly link chronic skin inflammation and sleep abnormalities characteristic of AD. The objective of this work is to systematically present and summarize the results of studies (both experimental and clinical) that investigated the role of MT in the AD, with a focus on the antioxidant and immunomodulatory effects of MT.
Collapse
Affiliation(s)
- Andrzej Kazimierz Jaworek
- Department of Dermatology, Jagiellonian University Medical College, 31-501 Cracow, Poland;
- Correspondence:
| | - Jacek Cezary Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Przemysław Hałubiec
- Student Scientific Group, Department of Dermatology, Jagiellonian University Medical College, 31-501 Cracow, Poland;
| | - Anna Wojas-Pelc
- Department of Dermatology, Jagiellonian University Medical College, 31-501 Cracow, Poland;
| | - Jolanta Jaworek
- Department of Medical Physiology, Faculty of Health Sciences, Jagiellonian University Medical College, 31-126 Cracow, Poland;
| |
Collapse
|
284
|
Huang C, Santofimia-Castaño P, Iovanna J. NUPR1: A Critical Regulator of the Antioxidant System. Cancers (Basel) 2021; 13:cancers13153670. [PMID: 34359572 PMCID: PMC8345110 DOI: 10.3390/cancers13153670] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Nuclear protein 1 (NUPR1) is activated in cellular stress and is expressed at high levels in cancer cells. Much evidence has been gathered supporting its critical role in regulating the antioxidant system. Our review aims to summarize the literature data on the impact of NUPR1 on the oxidative stress response via such a regulatory role and how its inhibition induces reactive oxygen species-mediated cell death, such as ferroptosis. Abstract Nuclear protein 1 (NUPR1) is a small intrinsically disordered protein (IDP) activated in response to various types of cellular stress, including endoplasmic reticulum (ER) stress and oxidative stress. Reactive oxygen species (ROS) are mainly produced during mitochondrial oxidative metabolism, and directly impact redox homeostasis and oxidative stress. Ferroptosis is a ROS-dependent programmed cell death driven by an iron-mediated redox reaction. Substantial evidence supports a maintenance role of the stress-inducible protein NUPR1 on cancer cell metabolism that confers chemotherapeutic resistance by upregulating mitochondrial function-associated genes and various antioxidant genes in cancer cells. NUPR1, identified as an antagonist of ferroptosis, plays an important role in redox reactions. This review summarizes the current knowledge on the mechanism behind the observed impact of NUPR1 on mitochondrial function, energy metabolism, iron metabolism, and the antioxidant system. The therapeutic potential of genetic or pharmacological inhibition of NUPR1 in cancer is also discussed. Understanding the role of NUPR1 in the antioxidant system and the mechanisms behind its regulation of ferroptosis may promote the development of more efficacious strategies for cancer therapy.
Collapse
|
285
|
Li X, Lv J, Li J, Ren X. Kir4.1 may represent a novel therapeutic target for diabetic retinopathy (Review). Exp Ther Med 2021; 22:1021. [PMID: 34373707 PMCID: PMC8343704 DOI: 10.3892/etm.2021.10453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022] Open
Abstract
As the major cause of irreversible loss of vision in adults, diabetic retinopathy (DR) is one of the most serious complications of diabetes. The imbalance of the retinal microenvironment and destruction of the blood-retinal barrier have a significant role in the progression of DR. Inward rectifying potassium channel 4.1 (Kir4.1) is located on Müller cells and is closely related to potassium homeostasis, water balance and glutamate clearance in the whole retina. The present review discusses the functions of Kir4.1 in regulating the retinal microenvironment and related biological mechanisms in DR. In the future, Kir4.1 may represent a novel alternative therapeutic target for DR through affecting the retinal microenvironment.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Department of Radiotherapy Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Jiajun Lv
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Department of Radiotherapy Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Jiazhi Li
- Department of Radiotherapy Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Xiang Ren
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
286
|
Santi C, Scimmi C, Sancineto L. Ebselen and Analogues: Pharmacological Properties and Synthetic Strategies for Their Preparation. Molecules 2021; 26:4230. [PMID: 34299505 PMCID: PMC8306772 DOI: 10.3390/molecules26144230] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Ebselen is the leader of selenorganic compounds, and starting from its identification as mimetic of the key antioxidant enzyme glutathione peroxidase, several papers have appeared in literature claiming its biological activities. It was the subject of several clinical trials and it is currently in clinical evaluation for the treatment of COVID-19 patients. Given our interest in the synthesis and pharmacological evaluation of selenorganic derivatives with this review, we aimed to collect all the papers focused on the biological evaluation of ebselen and its close analogues, covering the timeline between 2016 and most of 2021. Our analysis evidences that, even if it lacks specificity when tested in vitro, being able to bind to every reactive cysteine, it proved to be always well tolerated in vivo, exerting no sign of toxicity whatever the administered doses. Besides, looking at the literature, we realized that no review article dealing with the synthetic approaches for the construction of the benzo[d][1,2]-selenazol-3(2H)-one scaffold is available; thus, a section of the present review article is completely devoted to this specific topic.
Collapse
Affiliation(s)
| | | | - Luca Sancineto
- Group of Catalysis and Green Organic Chemistry, Department of Pharmaceutical Sciences, University of Perugia Via del Liceo 1, 06122 Perugia, Italy; (C.S.); (C.S.)
| |
Collapse
|
287
|
Chen B, Xu X, Lin DD, Chen X, Xu YT, Liu X, Dong WG. KRT18 Modulates Alternative Splicing of Genes Involved in Proliferation and Apoptosis Processes in Both Gastric Cancer Cells and Clinical Samples. Front Genet 2021; 12:635429. [PMID: 34290732 PMCID: PMC8287183 DOI: 10.3389/fgene.2021.635429] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
Keratin 18 (KRT18), one of the most abundant keratins in epithelial and endothelial cells, has been reported to be aberrantly expressed in many malignancies and extensively regarded as a biomarker and important regulator in multiple cancers, including gastric cancer (GC). But the molecular regulatory mechanisms of KRT18 in GC patients and cells are largely unknown. In the present study, we analyzed the expression level of KRT18 in 450 stomach adenocarcinoma tissue samples from TCGA database and found a significantly higher expression level in tumor tissues. We then explored the potential functions of KRT18 in AGS cells (human gastric adenocarcinoma cell line) by KRT18 knockdown using siRNA and whole transcriptome RNA-seq analysis. Notably, KRT18 selectively regulates expression of cell proliferation and apoptotic genes. Beyond this, KRT18 affects the alternative splicing of genes enriched in apoptosis, cell cycle, and other cancer-related pathways, which were then validated by reverse transcription-quantitative polymerase chain reaction approach. We validated KRT18-KD promoted apoptosis and inhibited proliferation in AGS cells. We then used RNA-seq data of GC samples to further demonstrate the modulation of KRT18 on alternative splicing regulation. These results together support the conclusion that KRT18 extensively modulates diverse alternative splicing events of genes enriched in proliferation and apoptosis processes. And the dysregulated splicing factors at transcriptional or posttranscriptional level by KRT18 may contribute to the alternative splicing change of many genes, which expands the functional importance of keratins in apoptotic and cell cycle pathways at the posttranscriptional level in GC.
Collapse
Affiliation(s)
- Biao Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ximing Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dan-dan Lin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang-tao Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei-guo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
288
|
Villavicencio Tejo F, Quintanilla RA. Contribution of the Nrf2 Pathway on Oxidative Damage and Mitochondrial Failure in Parkinson and Alzheimer's Disease. Antioxidants (Basel) 2021; 10:1069. [PMID: 34356302 PMCID: PMC8301100 DOI: 10.3390/antiox10071069] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 12/17/2022] Open
Abstract
The increase in human life expectancy has become a challenge to reduce the deleterious consequences of aging. Nowadays, an increasing number of the population suffer from age-associated neurodegenerative diseases including Parkinson's disease (PD) and Alzheimer's disease (AD). These disorders present different signs of neurodegeneration such as mitochondrial dysfunction, inflammation, and oxidative stress. Accumulative evidence suggests that the transcriptional factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) plays a vital defensive role orchestrating the antioxidant response in the brain. Nrf2 activation promotes the expression of several antioxidant enzymes that exert cytoprotective effects against oxidative damage and mitochondrial impairment. In this context, several studies have proposed a role of Nrf2 in the pathogenesis of PD and AD. Thus, we consider it important to summarize the ongoing literature related to the effects of the Nrf2 pathway in the context of these diseases. Therefore, in this review, we discuss the mechanisms involved in Nrf2 activity and its connection with mitochondria, energy supply, and antioxidant response in the brain. Furthermore, we will lead our discussion to identify the participation of the Nrf2 pathway in mitochondrial impairment and neurodegeneration present in PD and AD. Finally, we will discuss the therapeutic effects that the Nrf2 pathway activation could have on the cognitive impairment, neurodegeneration, and mitochondrial failure present in PD and AD.
Collapse
Affiliation(s)
| | - Rodrigo A Quintanilla
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910060, Chile;
| |
Collapse
|
289
|
Yip HK, Dubey NK, Lin KC, Sung PH, Chiang JY, Chu YC, Huang CR, Chen YL, Deng YH, Cheng HC, Deng WP. Melatonin rescues cerebral ischemic events through upregulated tunneling nanotube-mediated mitochondrial transfer and downregulated mitochondrial oxidative stress in rat brain. Biomed Pharmacother 2021; 139:111593. [PMID: 33865018 DOI: 10.1016/j.biopha.2021.111593] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Cerebral ischemic events, comprising of excitotoxicity, reactive oxygen production, and inflammation, adversely impact the metabolic-redox circuit in highly active neuronal metabolic profile which maintains energy-dependent brain activities. Therefore, we investigated neuro-regenerative potential of melatonin (Mel), a natural biomaterial secreted by pineal gland. METHODS We specifically determined whether Mel could influence tunneling nanotubes (TNTs)-mediated transfer of functional mitochondria (Mito) which in turn may alter membrane potential, oxidative stress and apoptotic factors. In vitro studies assessed the effects of Mito on levels of cytochrome C, mitochondrial transfer, reactive oxygen species, membrane potential and mass, which were all further enhanced by Mel pre-treatment, whereas in vivo studies examined brain infarct area (BIA), neurological function, inflammation, brain edema and integrity of neurons and myelin sheath in control, ischemia stroke (IS), IS + Mito and IS + Mel-Mito group rats. RESULTS Results showed that Mel pre-treatment significantly increased mitochondrial transfer and antioxidants, and inhibited apoptosis. Mel-pretreated Mito also significantly reduced BIA with improved neurological function. Apoptotic, oxidative-stress, autophagic, mitochondrial/DNA-damaged biomarkers indices were also improved. CONCLUSION Conclusively, Mel is a potent biomaterial which could potentially impart neurogenesis through repairing impaired metabolic-redox circuit via enhanced TNT-mediated mitochondrial transfer, anti-oxidation, and anti-apoptotic activities in ischemia.
Collapse
Affiliation(s)
- Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan; Department of Nursing, Asia University, Taichung 41354, Taiwan; Division of Cardiology, Department of Internal Medicine, Xiamen Chang Gung Hospital, Xiamen, Fujian 361000, China
| | - Navneet Kumar Dubey
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Kun-Chen Lin
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Pei-Hsun Sung
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - John Y Chiang
- Department of Computer Science and Engineering, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yi-Ching Chu
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chi-Ruei Huang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Yi-Ling Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Yue-Hua Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsin-Chung Cheng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Dentistry, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Win-Ping Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Basic Medicine, Fu Jen Catholic University, Taipei, Taiwan; Department of Life Science, Tunghai University, Taichung, Taiwan.
| |
Collapse
|
290
|
Fenton-Navarro B, Garduño Ríos D, Torner L, Letechipía-Vallejo G, Cervantes M. Melatonin Decreases Circulating Levels of Galectin-3 and Cytokines, Motor Activity, and Anxiety Following Acute Global Cerebral Ischemia in Male Rats. Arch Med Res 2021; 52:505-513. [DOI: 10.1016/j.arcmed.2021.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/21/2020] [Accepted: 01/21/2021] [Indexed: 12/23/2022]
|
291
|
Borrello MT, Santofimia-Castaño P, Bocchio M, Listi A, Fraunhoffer N, Soubeyran P, Chevet E, Pin C, Iovanna J. NUPR1 interacts with eIF2α and is required for resolution of the ER stress response in pancreatic tissue. FEBS J 2021; 288:4081-4097. [PMID: 33403797 DOI: 10.1111/febs.15700] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 11/20/2020] [Accepted: 01/04/2021] [Indexed: 12/22/2022]
Abstract
Nuclear protein 1 (NUPR1) is a stress response protein overexpressed upon cell injury in virtually all organs including the exocrine pancreas. Despite NUPR1's well-established role in the response to cell stress, the molecular and structural machineries triggered by NUPR1 activation remain largely debated. In this study, we uncover a new role for NUPR1, participating in the unfolded protein response (UPR) and the integrated stress response. Biochemical results and ultrastructural morphological observations revealed alterations in the UPR of acinar cells of germline-deleted NUPR1 murine models, consistent with the inability to restore general protein synthesis after stress induction. Bioinformatic analysis of NUPR1-interacting partners showed significant enrichment in translation initiation factors, including eukaryotic initiation factor (eIF) 2α. Co-immunoprecipitation and proximity ligation assays confirmed the interaction between NUPR1 and eIF2α and its phosphorylated form (p-eIF2α). Furthermore, our data suggest loss of NUPR1 in cells results in maintained eIF2α phosphorylation and evaluation of nascent proteins by click chemistry revealed that NUPR1-depleted PANC-1 cells displayed a slower poststress protein synthesis recovery when compared to wild-type. Combined, these data propose a novel role for NUPR1 in the integrated stress response pathway, at least partially through promoting efficient PERK branch activity and resolution through a unique interaction with eIF2α.
Collapse
Affiliation(s)
- Maria Teresa Borrello
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Marco Bocchio
- INMED (INSERM U1249), Turing Center for Living Systems, Aix-Marseille University, Marseille, France
| | - Angela Listi
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Nicolas Fraunhoffer
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Philippe Soubeyran
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Eric Chevet
- INSERM U1242, Proteostasis and Cancer Team, Chemistry Oncogenesis Stress Signaling, Université de Rennes 1, Rennes, France
| | - Christopher Pin
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
- Departments of Pediatrics, Oncology, and Physiology and Pharmacology, Schulich School of Medicine, University of Western Ontario, Children's Health Research Institute, London, ON, Canada
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
292
|
Hernández IH, Villa-González M, Martín G, Soto M, Pérez-Álvarez MJ. Glial Cells as Therapeutic Approaches in Brain Ischemia-Reperfusion Injury. Cells 2021; 10:1639. [PMID: 34208834 PMCID: PMC8305833 DOI: 10.3390/cells10071639] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke is the second cause of mortality and the first cause of long-term disability constituting a serious socioeconomic burden worldwide. Approved treatments include thrombectomy and rtPA intravenous administration, which, despite their efficacy in some cases, are not suitable for a great proportion of patients. Glial cell-related therapies are progressively overcoming inefficient neuron-centered approaches in the preclinical phase. Exploiting the ability of microglia to naturally switch between detrimental and protective phenotypes represents a promising therapeutic treatment, in a similar way to what happens with astrocytes. However, the duality present in many of the roles of these cells upon ischemia poses a notorious difficulty in disentangling the precise pathways to target. Still, promoting M2/A2 microglia/astrocyte protective phenotypes and inhibiting M1/A1 neurotoxic profiles is globally rendering promising results in different in vivo models of stroke. On the other hand, described oligodendrogenesis after brain ischemia seems to be strictly beneficial, although these cells are the less studied players in the stroke paradigm and negative effects could be described for oligodendrocytes in the next years. Here, we review recent advances in understanding the precise role of mentioned glial cell types in the main pathological events of ischemic stroke, including inflammation, blood brain barrier integrity, excitotoxicity, reactive oxygen species management, metabolic support, and neurogenesis, among others, with a special attention to tested therapeutic approaches.
Collapse
Affiliation(s)
- Ivó H Hernández
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Mario Villa-González
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Gerardo Martín
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Manuel Soto
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - María José Pérez-Álvarez
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
293
|
El-Sikaily A, Helal M. Environmental pollution and diabetes mellitus. World J Meta-Anal 2021; 9:234-256. [DOI: 10.13105/wjma.v9.i3.234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/17/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus (DM) is a chromic metabolic disease that affects a large segment of the population worldwide. Physical inactivity, poor nutrition, and genetic predisposition are main risk factors for disease development. In the last decade, it was clear to the scientific community that DM development is linked to a novel disease inducer that was later defined as diabetogenic factors of pollution and endocrine disrupting agents. Environmental pollution is exponentially increasing in uncontrolled manner in several countries. Environmental pollutants are of diverse nature and toxicities, including polyaromatic hydrocarbons (PAHs), pesticides, and heavy metals. In the current review, we shed light on the impact of each class of these pollutants and the underlined molecular mechanism of diabetes induction and biological toxicities. Finally, a brief overview about the connection between coronavirus disease 2019 and diabetes pandemics is presented.
Collapse
Affiliation(s)
- Amany El-Sikaily
- National Institute of Oceanography and Fisheries (NIOF), Cairo 21513, Egypt
| | - Mohamed Helal
- National Institute of Oceanography and Fisheries (NIOF), Cairo 21513, Egypt
| |
Collapse
|
294
|
RGS14 Regulation of Post-Synaptic Signaling and Spine Plasticity in Brain. Int J Mol Sci 2021; 22:ijms22136823. [PMID: 34201943 PMCID: PMC8268017 DOI: 10.3390/ijms22136823] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023] Open
Abstract
The regulator of G-protein signaling 14 (RGS14) is a multifunctional signaling protein that regulates post synaptic plasticity in neurons. RGS14 is expressed in the brain regions essential for learning, memory, emotion, and stimulus-induced behaviors, including the basal ganglia, limbic system, and cortex. Behaviorally, RGS14 regulates spatial and object memory, female-specific responses to cued fear conditioning, and environmental- and psychostimulant-induced locomotion. At the cellular level, RGS14 acts as a scaffolding protein that integrates G protein, Ras/ERK, and calcium/calmodulin signaling pathways essential for spine plasticity and cell signaling, allowing RGS14 to naturally suppress long-term potentiation (LTP) and structural plasticity in hippocampal area CA2 pyramidal cells. Recent proteomics findings indicate that RGS14 also engages the actomyosin system in the brain, perhaps to impact spine morphogenesis. Of note, RGS14 is also a nucleocytoplasmic shuttling protein, where its role in the nucleus remains uncertain. Balanced nuclear import/export and dendritic spine localization are likely essential for RGS14 neuronal functions as a regulator of synaptic plasticity. Supporting this idea, human genetic variants disrupting RGS14 localization also disrupt RGS14’s effects on plasticity. This review will focus on the known and unexplored roles of RGS14 in cell signaling, physiology, disease and behavior.
Collapse
|
295
|
Estévez M. Critical overview of the use of plant antioxidants in the meat industry: Opportunities, innovative applications and future perspectives. Meat Sci 2021; 181:108610. [PMID: 34147961 DOI: 10.1016/j.meatsci.2021.108610] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/19/2022]
Abstract
The number of articles devoted to study the effect of "natural antioxidants" on meat systems has remarkably increased in the last 10 years. Yet, a critical review of literature reveals recurrent flaws in regards to the rationale of the application, the experimental design, the characterisation of the plant sources, the discussion of the molecular mechanisms and of the potential benefits. The selection of the appropriate source of these antioxidants and the identification of their bioactive constituents, are essential to understand their mode of action and set effective and safe doses. The methodological approach should also be planned with care as the recorded effects and main conclusions largely depend on the accuracy and specificity of the methods. This article aims to critically review the recent advances in the application of plant antioxidants in meat and meat products and briefly covers current trends of innovative application and future trends.
Collapse
Affiliation(s)
- M Estévez
- Meat and Meat Products Research Institute (IPROCAR), Food Technology, University of Extremadura, 10003 Cáceres, Spain.
| |
Collapse
|
296
|
Chuai H, Zhang SQ, Bai H, Li J, Wang Y, Sun J, Wen E, Zhang J, Xin M. Small molecule selenium-containing compounds: Recent development and therapeutic applications. Eur J Med Chem 2021; 223:113621. [PMID: 34217061 DOI: 10.1016/j.ejmech.2021.113621] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023]
Abstract
Selenium (Se) is an essential micronutrient of organism and has important function. It participates in the functions of selenoprotein in several manners. In recent years, Se has attracted much attention because of its therapeutic potential against several diseases. Many natural and synthetic organic Se-containing compounds were studied and explored for the treatment of cancer and other diseases. Studies have showed that incorporation of Se atom into small molecules significantly enhanced their bioactivities. In this paper, according to different applications and structural characteristics, the research progress and therapeutic application of Se-containing compounds are reviewed, and more than 110 Se-containing compounds were selected as representatives which showed potent activities such as anticancer, antioxidant, antifibrolytic, antiparasitic, antibacterial, antiviral, antifungal and central nervous system related effects. This review is expected to provide a basis for further study of new promising Se-containing compounds.
Collapse
Affiliation(s)
- Hongyan Chuai
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - San-Qi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - Huanrong Bai
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - Jiyu Li
- Henan Xibaikang Health Industry Co., Ltd, Jiyuan, Henan, 459006, PR China
| | - Yang Wang
- Henan Xibaikang Health Industry Co., Ltd, Jiyuan, Henan, 459006, PR China
| | - Jiajia Sun
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - Ergang Wen
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - Jiye Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - Minhang Xin
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China.
| |
Collapse
|
297
|
Vermot A, Petit-Härtlein I, Smith SME, Fieschi F. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology. Antioxidants (Basel) 2021; 10:890. [PMID: 34205998 PMCID: PMC8228183 DOI: 10.3390/antiox10060890] [Citation(s) in RCA: 362] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 01/17/2023] Open
Abstract
The reactive oxygen species (ROS)-producing enzyme NADPH oxidase (NOX) was first identified in the membrane of phagocytic cells. For many years, its only known role was in immune defense, where its ROS production leads to the destruction of pathogens by the immune cells. NOX from phagocytes catalyzes, via one-electron trans-membrane transfer to molecular oxygen, the production of the superoxide anion. Over the years, six human homologs of the catalytic subunit of the phagocyte NADPH oxidase were found: NOX1, NOX3, NOX4, NOX5, DUOX1, and DUOX2. Together with the NOX2/gp91phox component present in the phagocyte NADPH oxidase assembly itself, the homologs are now referred to as the NOX family of NADPH oxidases. NOX are complex multidomain proteins with varying requirements for assembly with combinations of other proteins for activity. The recent structural insights acquired on both prokaryotic and eukaryotic NOX open new perspectives for the understanding of the molecular mechanisms inherent to NOX regulation and ROS production (superoxide or hydrogen peroxide). This new structural information will certainly inform new investigations of human disease. As specialized ROS producers, NOX enzymes participate in numerous crucial physiological processes, including host defense, the post-translational processing of proteins, cellular signaling, regulation of gene expression, and cell differentiation. These diversities of physiological context will be discussed in this review. We also discuss NOX misregulation, which can contribute to a wide range of severe pathologies, such as atherosclerosis, hypertension, diabetic nephropathy, lung fibrosis, cancer, or neurodegenerative diseases, giving this family of membrane proteins a strong therapeutic interest.
Collapse
Affiliation(s)
- Annelise Vermot
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| | - Isabelle Petit-Härtlein
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| | - Susan M. E. Smith
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA;
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| |
Collapse
|
298
|
Wang J, Struebing FL, Geisert EE. Commonalities of optic nerve injury and glaucoma-induced neurodegeneration: Insights from transcriptome-wide studies. Exp Eye Res 2021; 207:108571. [PMID: 33844961 PMCID: PMC9890784 DOI: 10.1016/j.exer.2021.108571] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/28/2021] [Accepted: 04/02/2021] [Indexed: 02/03/2023]
Abstract
Glaucoma is a collection of diseases that lead to an irreversible vision loss due to damage of retinal ganglion cells (RGCs). Although the underlying events leading to RGC death are not fully understood, recent research efforts are beginning to define the genetic changes that play a critical role in the initiation and progression of glaucomatous injury and RGC death. Several genetic and experimental animal models have been developed to mimic glaucomatous neurodegeneration. These models differ in many respects but all result in the loss of RGCs. Assessing transcriptional changes across different models could provide a more complete perspective on the molecular drivers of RGC degeneration. For the past several decades, changes in the retinal transcriptome during neurodegeneration process were defined using microarray methods, RNA sequencing and now single cell RNA sequencing. It is understood that these methods have strengths and weaknesses due to technical differences and variations in the analytical tools used. In this review, we focus on the use of transcriptome-wide expression profiling of the changes occurring as RGCs are lost across different glaucoma models. Commonalities of optic nerve crush and glaucoma-induced neurodegeneration are identified and discussed.
Collapse
Affiliation(s)
- Jiaxing Wang
- Emory Eye Center, Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA
| | - Felix L. Struebing
- Center for Neuropathology and Prion Research, Ludwig Maximilian University of Munich, Germany,Department for Translational Brain Research, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Eldon E. Geisert
- Emory Eye Center, Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA,Corresponding author: (E.E. Geisert)
| |
Collapse
|
299
|
Estaras M, Gonzalez-Portillo MR, Fernandez-Bermejo M, Mateos JM, Vara D, Blanco-Fernandez G, Lopez-Guerra D, Roncero V, Salido GM, González A. Melatonin Induces Apoptosis and Modulates Cyclin Expression and MAPK Phosphorylation in Pancreatic Stellate Cells Subjected to Hypoxia. Int J Mol Sci 2021; 22:5555. [PMID: 34074034 PMCID: PMC8197391 DOI: 10.3390/ijms22115555] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023] Open
Abstract
In certain diseases of the pancreas, pancreatic stellate cells form an important part of fibrosis and are critical for the development of cancer cells. A hypoxic condition develops within the tumor, to which pancreatic stellate cells adapt and are able to proliferate. The consequence is the growth of the tumor. Melatonin, the product of the pineal gland, is gaining attention as an agent with therapeutic potential against pancreatic cancers. Its actions on tumor cells lead, in general, to a reduction in cell viability and proliferation. However, its effects on pancreatic stellate cells subjected to hypoxia are less known. In this study, we evaluated the actions of pharmacological concentrations of melatonin (1 mM-1 µM) on pancreatic stellate cells subjected to hypoxia. The results show that melatonin induced a decrease in cell viability at the highest concentrations tested. Similarly, the incorporation of BrdU into DNA was diminished by melatonin. The expression of cyclins A and D also was decreased in the presence of melatonin. Upon treatment of cells with melatonin, increases in the expression of major markers of ER stress, namely BIP, phospho-eIF2α and ATF-4, were detected. Modulation of apoptosis was noticed as an increase in caspase-3 activation. In addition, changes in the phosphorylated state of p44/42, p38 and JNK MAPKs were detected in cells treated with melatonin. A slight decrease in the content of α-smooth muscle actin was detected in cells treated with melatonin. Finally, treatment of cells with melatonin decreased the expression of matrix metalloproteinases 2, 3, 9 and 13. Our observations suggest that melatonin, at pharmacological concentrations, diminishes the proliferation of pancreatic stellate cells subjected to hypoxia through modulation of cell cycle, apoptosis and the activation of crucial MAPKs. Cellular responses might involve certain ER stress regulator proteins. In view of the results, melatonin could be taken into consideration as a potential therapeutic agent for pancreatic fibrosis.
Collapse
Affiliation(s)
- Matias Estaras
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| | - Manuel R. Gonzalez-Portillo
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| | - Miguel Fernandez-Bermejo
- Department of Gastroenterology, San Pedro de Alcantara Hospital, 10003 Caceres, Spain; (M.F.-B.); (J.M.M.); (D.V.)
| | - Jose M. Mateos
- Department of Gastroenterology, San Pedro de Alcantara Hospital, 10003 Caceres, Spain; (M.F.-B.); (J.M.M.); (D.V.)
| | - Daniel Vara
- Department of Gastroenterology, San Pedro de Alcantara Hospital, 10003 Caceres, Spain; (M.F.-B.); (J.M.M.); (D.V.)
| | - Gerardo Blanco-Fernandez
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, University Hospital, 06080 Badajoz, Spain; (G.B.-F.); (D.L.-G.)
| | - Diego Lopez-Guerra
- Hepatobiliary-Pancreatic Surgery and Liver Transplant Unit, University Hospital, 06080 Badajoz, Spain; (G.B.-F.); (D.L.-G.)
| | - Vicente Roncero
- Unit of Histology and Pathological Anatomy, Veterinary Faculty, University of Extremadura, 10003 Caceres, Spain;
| | - Gines M. Salido
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| | - Antonio González
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (M.E.); (M.R.G.-P.); (G.M.S.)
| |
Collapse
|
300
|
Liu T, Lv YF, Zhao JL, You QD, Jiang ZY. Regulation of Nrf2 by phosphorylation: Consequences for biological function and therapeutic implications. Free Radic Biol Med 2021; 168:129-141. [PMID: 33794311 DOI: 10.1016/j.freeradbiomed.2021.03.034] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/18/2022]
Abstract
The transcription factor nuclear factor erythroid-derived 2-like 2 (NRF2) participates in the activation of the antioxidant cytoprotective pathway and other important physiological processes to maintain cellular homeostasis. The dysregulation of NRF2 activity plays a role in various diseases, such as cardiovascular diseases, neurodegenerative diseases, and cancer. Thus, NRF2 activity is tightly regulated through multiple mechanisms, among which phosphorylation by kinases is critical in the posttranslational regulation of NRF2. For instance, PKC, casein kinase 2, and AMP-activated kinase positively, while GSK-3 negatively regulates NRF2 activity through phosphorylation of different sites. Here, we provide an overview of the phosphorylation regulation pattern of NRF2 and discuss the therapeutic potential of interventions targeting NRF2 phosphorylation.
Collapse
Affiliation(s)
- Tian Liu
- State Key Laboratory of Natural Medicines, And Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
| | - Yi-Fei Lv
- State Key Laboratory of Natural Medicines, And Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
| | - Jing-Long Zhao
- State Key Laboratory of Natural Medicines, And Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
| | - Qi-Dong You
- State Key Laboratory of Natural Medicines, And Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Zheng-Yu Jiang
- State Key Laboratory of Natural Medicines, And Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|