1
|
Czajkowski K, Herbet M, Murias M, Piątkowska-Chmiel I. Senolytics: charting a new course or enhancing existing anti-tumor therapies? Cell Oncol (Dordr) 2025; 48:351-371. [PMID: 39633108 PMCID: PMC11996976 DOI: 10.1007/s13402-024-01018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Cell senescence is a natural response within our organisms. Initially, it was considered an effective anti-tumor mechanism. However, it is now believed that while cell senescence initially acts as a robust barrier against tumor initiation, the subsequent accumulation of senescent cells can paradoxically promote cancer recurrence and cause damage to neighboring tissues. This intricate balance between cell proliferation and senescence plays a pivotal role in maintaining tissue homeostasis. Moreover, senescence cells secrete many bioactive molecules collectively termed the senescence-associated secretory phenotype (SASP), which can induce chronic inflammation, alter tissue architecture, and promote tumorigenesis through paracrine signaling. Among the myriads of compounds, senotherapeutic drugs have emerged as exceptionally promising candidates in anticancer treatment. Their ability to selectively target senescent cells while sparing healthy tissues represents a paradigm shift in therapeutic intervention, offering new avenues for personalized oncology medicine. Senolytics have introduced new therapeutic possibilities by enabling the targeted removal of senescent cells. As standalone agents, they can clear tumor cells in a senescent state and, when combined with chemo- or radiotherapy, eliminate residual senescent cancer cells after treatment. This dual approach allows for the intentional use of lower-dose therapies or the removal of unintended senescent cells post-treatment. Additionally, by targeting non-cancerous senescent cells, senolytics may help reduce tumor formation risk, limit recurrence, and slow disease progression. This article examines the mechanisms of cellular senescence, its role in cancer treatment, and the importance of senotherapy, with particular attention to the therapeutic potential of senolytic drugs.
Collapse
Affiliation(s)
- Konrad Czajkowski
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Mariola Herbet
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Marek Murias
- Department of Toxicology, Poznan University of Medical Sciences, Poznań, Poland
| | - Iwona Piątkowska-Chmiel
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
2
|
Komarudin AG, Adharis A, Sasmono RT. Natural Compounds and Their Analogs as Antivirals Against Dengue Virus: A Review. Phytother Res 2025; 39:888-921. [PMID: 39697048 DOI: 10.1002/ptr.8408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 12/20/2024]
Abstract
Dengue virus (DENV) continues to pose a significant global health challenge, causing diseases such as dengue fever, dengue hemorrhagic fever, and dengue shock syndrome. While efforts in vaccine development and antiviral drug discovery are ongoing, effective therapeutic options remain limited. In this review, we highlight natural compounds and the analogs that demonstrated antiviral activity against DENV in in vitro and in vivo studies. Specifically, these studies examine alkaloids, phenolic acids, phenols, flavonoids, terpenoids, and glycosides which have shown potential in inhibiting DENV entry, replication, and reducing the cytokine storm. By focusing on these bioactive compounds and the analogs, a comprehensive overview of their promising roles is provided to advance therapeutic strategies for combating DENV infection.
Collapse
Affiliation(s)
- Amalina Ghaisani Komarudin
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency (BRIN), Kabupaten Bogor, Jawa Barat, Indonesia
| | - Azis Adharis
- Department of Chemistry, Faculty of Science and Computer Science, Universitas Pertamina (UPER), Jakarta, Indonesia
| | - R Tedjo Sasmono
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency (BRIN), Kabupaten Bogor, Jawa Barat, Indonesia
| |
Collapse
|
3
|
Banse SA, Sedore CA, Johnson E, Coleman-Hulbert AL, Onken B, Hall D, Jackson EG, Huynh P, Foulger AC, Guo S, Garrett T, Xue J, Inman D, Morshead ML, Plummer WT, Chen E, Bhaumik D, Chen MK, Harinath G, Chamoli M, Quinn RP, Falkowski R, Edgar D, Schmidt MO, Lucanic M, Guo M, Driscoll M, Lithgow GJ, Phillips PC. Antioxidants green tea extract and nordihydroguaiaretic acid confer species and strain-specific lifespan and health effects in Caenorhabditis nematodes. GeroScience 2024; 46:2239-2251. [PMID: 37923874 PMCID: PMC10828308 DOI: 10.1007/s11357-023-00978-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/08/2023] [Indexed: 11/06/2023] Open
Abstract
The Caenorhabditis Intervention Testing Program (CITP) is an NIH-funded research consortium of investigators who conduct analyses at three independent sites to identify chemical interventions that reproducibly promote health and lifespan in a robust manner. The founding principle of the CITP is that compounds with positive effects across a genetically diverse panel of Caenorhabditis species and strains are likely engaging conserved biochemical pathways to exert their effects. As such, interventions that are broadly efficacious might be considered prominent compounds for translation for pre-clinical research and human clinical applications. Here, we report results generated using a recently streamlined pipeline approach for the evaluation of the effects of chemical compounds on lifespan and health. We studied five compounds previously shown to extend C. elegans lifespan or thought to promote mammalian health: 17α-estradiol, acarbose, green tea extract, nordihydroguaiaretic acid, and rapamycin. We found that green tea extract and nordihydroguaiaretic acid extend Caenorhabditis lifespan in a species-specific manner. Additionally, these two antioxidants conferred assay-specific effects in some studies-for example, decreasing survival for certain genetic backgrounds in manual survival assays in contrast with extended lifespan as assayed using automated C. elegans Lifespan Machines. We also observed that GTE and NDGA impact on older adult mobility capacity is dependent on genetic background, and that GTE reduces oxidative stress resistance in some Caenorhabditis strains. Overall, our analysis of the five compounds supports the general idea that genetic background and assay type can influence lifespan and health effects of compounds, and underscores that lifespan and health can be uncoupled by chemical interventions.
Collapse
Affiliation(s)
- Stephen A Banse
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR, 97403, USA
| | - Christine A Sedore
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR, 97403, USA
| | - Erik Johnson
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR, 97403, USA
| | | | - Brian Onken
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers University, Piscataway, NJ, 08854, USA
| | - David Hall
- The Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - E Grace Jackson
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR, 97403, USA
| | - Phu Huynh
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers University, Piscataway, NJ, 08854, USA
| | - Anna C Foulger
- The Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Suzhen Guo
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers University, Piscataway, NJ, 08854, USA
| | - Theo Garrett
- The Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Jian Xue
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers University, Piscataway, NJ, 08854, USA
| | - Delaney Inman
- The Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | | | - W Todd Plummer
- The Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Esteban Chen
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers University, Piscataway, NJ, 08854, USA
| | - Dipa Bhaumik
- The Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Michelle K Chen
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR, 97403, USA
| | - Girish Harinath
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers University, Piscataway, NJ, 08854, USA
| | - Manish Chamoli
- The Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Rose P Quinn
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR, 97403, USA
| | - Ron Falkowski
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers University, Piscataway, NJ, 08854, USA
| | - Daniel Edgar
- The Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Madeline O Schmidt
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR, 97403, USA
| | - Mark Lucanic
- The Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Max Guo
- Division of Aging Biology, National Institute On Aging, Bethesda, MD, 20892-9205, USA
| | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers University, Piscataway, NJ, 08854, USA.
| | - Gordon J Lithgow
- The Buck Institute for Research On Aging, Novato, CA, 94945, USA.
| | - Patrick C Phillips
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR, 97403, USA.
| |
Collapse
|
4
|
Osmakov DI, Kalinovskii AP, Belozerova OA, Andreev YA, Kozlov SA. Lignans as Pharmacological Agents in Disorders Related to Oxidative Stress and Inflammation: Chemical Synthesis Approaches and Biological Activities. Int J Mol Sci 2022; 23:6031. [PMID: 35682715 PMCID: PMC9181380 DOI: 10.3390/ijms23116031] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023] Open
Abstract
Plant lignans exhibit a wide range of biological activities, which makes them the research objects of potential use as therapeutic agents. They provide diverse naturally-occurring pharmacophores and are available for production by chemical synthesis. A large amount of accumulated data indicates that lignans of different structural groups are apt to demonstrate both anti-inflammatory and antioxidant effects, in many cases, simultaneously. In this review, we summarize the comprehensive knowledge about lignan use as a bioactive agent in disorders associated with oxidative stress and inflammation, pharmacological effects in vitro and in vivo, molecular mechanisms underlying these effects, and chemical synthesis approaches. This article provides an up-to-date overview of the current data in this area, available in PubMed, Scopus, and Web of Science databases, screened from 2000 to 2022.
Collapse
Affiliation(s)
- Dmitry I. Osmakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (D.I.O.); (A.P.K.); (O.A.B.); (Y.A.A.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Aleksandr P. Kalinovskii
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (D.I.O.); (A.P.K.); (O.A.B.); (Y.A.A.)
| | - Olga A. Belozerova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (D.I.O.); (A.P.K.); (O.A.B.); (Y.A.A.)
| | - Yaroslav A. Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (D.I.O.); (A.P.K.); (O.A.B.); (Y.A.A.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Sergey A. Kozlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (D.I.O.); (A.P.K.); (O.A.B.); (Y.A.A.)
| |
Collapse
|
5
|
Mala John GS, Takeuchi S, Venkatraman G, Rayala SK. Nordihydroguaiaretic Acid in Therapeutics: Beneficial to Toxicity Profiles and the Search for its Analogs. Curr Cancer Drug Targets 2021; 20:86-103. [PMID: 31642411 DOI: 10.2174/1568009619666191022141547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/26/2019] [Accepted: 08/22/2019] [Indexed: 12/16/2022]
Abstract
Nordihydroguaiaretic acid (NDGA) is a plant lignan obtained from creosote bush, Larrea tridentata and is known to possess antioxidant, anticancer activities and is used in traditional medicine in North America and Mexico. However, its prolonged consumption leads to liver damage and kidney dysfunction. Despite its toxicity and side effects, there is little awareness to forbid its consumption and its use in the treatment of medical ailments has continued over the years. Several reports discuss its therapeutic efficiency and its medical applications have tremendously been on the rise to date. There has been a recent surge of interest in the chemical synthesis of NDGA derivatives for therapeutic applications. NDGA derivatives have been developed as better alternatives to NDGA. Although several NDGA derivatives have been chemically synthesized as evidenced by recent literature, there is a paucity of information on their therapeutic efficacies. This review is to highlight the medicinal applications of NDGA, its toxicity evaluations and discuss the chemical derivatives of NDGA synthesized and studied so far and suggest to continue research interests in the development of NDGA analogs for therapeutic applications. We suggest that NDGA derivatives should be investigated more in terms of chemical synthesis with preferred conformational structures and exploit their biological potentials with future insights to explore in this direction to design and develop structurally modified NDGA derivatives for potential pharmacological properties.
Collapse
Affiliation(s)
| | - Satoru Takeuchi
- Factory of Takeuchi Nenshi, TAKENEN, 85NE Takamatsu, Kahoku Ishikawa 929-1215, Japan
| | - Ganesh Venkatraman
- Sri Ramachandra Center for Biomedical Nanotechnology, Sri Ramachandra Institute of Higher Education & Research, Chennai-600116, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology (IIT), Madras, Chennai-600036, India
| |
Collapse
|
6
|
Mongelli A, Atlante S, Barbi V, Bachetti T, Martelli F, Farsetti A, Gaetano C. Treating Senescence like Cancer: Novel Perspectives in Senotherapy of Chronic Diseases. Int J Mol Sci 2020; 21:ijms21217984. [PMID: 33121118 PMCID: PMC7663758 DOI: 10.3390/ijms21217984] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
The WHO estimated around 41 million deaths worldwide each year for age-related non-communicable chronic diseases. Hence, developing strategies to control the accumulation of cell senescence in living organisms and the overall aging process is an urgently needed problem of social relevance. During aging, many biological processes are altered, which globally induce the dysfunction of the whole organism. Cell senescence is one of the causes of this modification. Nowadays, several drugs approved for anticancer therapy have been repurposed to treat senescence, and others are under scrutiny in vitro and in vivo to establish their senomorphic or senolytic properties. In some cases, this research led to a significant increase in cell survival or to a prolonged lifespan in animal models, at least. Senomorphics can act to interfere with a specific pathway in order to restore the appropriate cellular function, preserve viability, and to prolong the lifespan. On the other hand, senolytics induce apoptosis in senescent cells allowing the remaining non–senescent population to preserve or restore tissue function. A large number of research articles and reviews recently addressed this topic. Herein, we would like to focus attention on those chemical agents with senomorphic or senolytic properties that perspectively, according to literature, suggest a potential application as senotherapeutics for chronic diseases.
Collapse
Affiliation(s)
- Alessia Mongelli
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy; (A.M.); (S.A.); (V.B.)
| | - Sandra Atlante
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy; (A.M.); (S.A.); (V.B.)
| | - Veronica Barbi
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy; (A.M.); (S.A.); (V.B.)
| | - Tiziana Bachetti
- Direzione Scientifica, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy;
| | - Fabio Martelli
- Laboratorio di Cardiologia Molecolare, Policlinico San Donato IRCCS, San Donato Milanese, 20097 Milano; Italy,
| | - Antonella Farsetti
- Institute for Systems Analysis and Computer Science “A. Ruberti” (IASI), National Research Council (CNR), 00185 Rome, Italy
- Correspondence: (A.F.); (C.G.)
| | - Carlo Gaetano
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy; (A.M.); (S.A.); (V.B.)
- Correspondence: (A.F.); (C.G.)
| |
Collapse
|
7
|
Manda G, Rojo AI, Martínez-Klimova E, Pedraza-Chaverri J, Cuadrado A. Nordihydroguaiaretic Acid: From Herbal Medicine to Clinical Development for Cancer and Chronic Diseases. Front Pharmacol 2020; 11:151. [PMID: 32184727 PMCID: PMC7058590 DOI: 10.3389/fphar.2020.00151] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 02/05/2020] [Indexed: 12/11/2022] Open
Abstract
Nordihydroguaiaretic acid (NDGA) is a phenolic lignan obtained from Larrea tridentata, the creosote bush found in Mexico and USA deserts, that has been used in traditional medicine for the treatment of numerous diseases such as cancer, renal, cardiovascular, immunological, and neurological disorders, and even aging. NDGA presents two catechol rings that confer a very potent antioxidant activity by scavenging oxygen free radicals and this may explain part of its therapeutic action. Additional effects include inhibition of lipoxygenases (LOXs) and activation of signaling pathways that impinge on the transcription factor Nuclear Factor Erythroid 2-related Factor (NRF2). On the other hand, the oxidation of the catechols to the corresponding quinones my elicit alterations in proteins and DNA that raise safety concerns. This review describes the current knowledge on NDGA, its targets and side effects, and its synthetic analogs as promising therapeutic agents, highlighting their mechanism of action and clinical projection towards therapy of neurodegenerative, liver, and kidney disease, as well as cancer.
Collapse
Affiliation(s)
- Gina Manda
- Department Cellular and Molecular Medicine, Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Ana I Rojo
- Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria la Paz (idiPAZ), Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, Spain
| | - Elena Martínez-Klimova
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Antonio Cuadrado
- Department Cellular and Molecular Medicine, Victor Babes National Institute of Pathology, Bucharest, Romania.,Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria la Paz (idiPAZ), Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, Spain
| |
Collapse
|
8
|
Han L, Bittner S, Dong D, Cortez Y, Dulay H, Arshad S, Shen WJ, Kraemer FB, Azhar S. Creosote bush-derived NDGA attenuates molecular and pathological changes in a novel mouse model of non-alcoholic steatohepatitis (NASH). Mol Cell Endocrinol 2019; 498:110538. [PMID: 31415794 PMCID: PMC7273809 DOI: 10.1016/j.mce.2019.110538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/26/2019] [Accepted: 08/11/2019] [Indexed: 02/06/2023]
Abstract
Creosote bush (Larrea tridentata)-derived nordihydroguaiaretic acid (NDGA) was shown to have profound effects on the core components of metabolic syndrome. This study investigated the in vivo potential of NDGA for prevention or attenuation of the pathophysiologic abnormalities of NASH. A novel dietary NASH model with feeding C57BL/6J mice with a high trans-fat, high cholesterol and high fructose (HTF) diet, was used. The HTF diet fed mice exhibited obesity, insulin resistance, hepatic steatosis, fibrosis, inflammation, ER stress, oxidative stress, and liver injury. NDGA attenuated these metabolic abnormalities as well as hepatic steatosis and fibrosis together with attenuated expression of genes encoding fibrosis, progenitor and macrophage markers with no effect on the levels of mRNAs for lipogenic enzymes. NDGA increased expression of fatty acid oxidation genes. In conclusion, NDGA exerts anti-NASH/anti-fibrotic actions and raises the therapeutic potential of NDGA for treatment of NASH patients with fibrosis and other associated complications.
Collapse
Affiliation(s)
- Lu Han
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA
| | - Stefanie Bittner
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA
| | - Dachuan Dong
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA
| | - Yuan Cortez
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA
| | - Hunter Dulay
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA
| | - Sara Arshad
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA
| | - Wen-Jun Shen
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA.
| | - Fredric B Kraemer
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, USA
| | - Salman Azhar
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, USA.
| |
Collapse
|
9
|
Singh M, Bittner S, Li Y, Bittner A, Han L, Cortez Y, Inayathullah M, Arif Z, Parthasarathi R, Rajadas J, Shen WJ, Nicolls MR, Kraemer FB, Azhar S. Anti-hyperlipidaemic effects of synthetic analogues of nordihydroguaiaretic acid in dyslipidaemic rats. Br J Pharmacol 2018; 176:369-385. [PMID: 30374952 DOI: 10.1111/bph.14528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 09/07/2018] [Accepted: 10/03/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Previous studies have shown that Creosote bush-derived nordihydroguaiaretic acid (NDGA) exerts beneficial actions on the key components of metabolic syndrome including dyslipidaemia, insulin resistance and hypertension in several relevant rodent models. Here, we synthesized and screened a total of 6 anti-hyperlipidaemic analogues of NDGA and tested their efficacy against hepatic lipid metabolism in a high-fructose diet (HFrD) fed dyslipidaemic rat model. EXPERIMENTAL APPROACH HFrD fed Sprague-Dawley rats treated with NDGA or one of the six analogues were used. Serum samples were analysed for blood metabolites, whereas liver samples were quantified for changes in various mRNA levels by real-time RT-PCR. KEY RESULTS Oral gavage of HFrD-fed rats for 4 days with NDGA analogues 1 and 2 (100 mg·kg-1 ·day-1 ) suppressed the hepatic triglyceride content, whereas the NDGA analogues 2, 3 and 4, like NDGA, decreased the plasma triglyceride levels by 70-75%. qRT-PCR measurements demonstrated that among NDGA analogues 1, 2, 4 and 5, analogue 4 was the most effective at inhibiting the mRNA levels of some key enzymes and transcription factors involved in lipogenesis. All four analogues almost equally inhibited the key genes involved in triglyceride synthesis and fatty acid elongation. Unlike NDGA, none of the analogues affected the genes of hepatic fatty acid oxidation or transport. CONCLUSIONS AND IMPLICATIONS Our data suggest that NDGA analogues 1, 2, 4 and 5, particularly analogue 4, exert their anti-hyperlipidaemic actions by negatively targeting genes of key enzymes and transcription factors involved in lipogenesis, triglyceride synthesis and fatty acid elongation. These analogues have therapeutic potential.
Collapse
Affiliation(s)
- Madhurima Singh
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| | - Stefanie Bittner
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Yihang Li
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| | - Alex Bittner
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Lu Han
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| | - Yuan Cortez
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | | | - Zeeshan Arif
- Computational Toxicology Facility, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | | | - Jayakumar Rajadas
- Division of Cardiovascular Pharmacology CVI, BioADD Laboratory, Stanford, CA, USA
| | - Wen-Jun Shen
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| | - Mark R Nicolls
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Fredric B Kraemer
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| | - Salman Azhar
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| |
Collapse
|
10
|
Liu Y, Zhang H, Yan L, Du W, Zhang M, Chen H, Zhang L, Li G, Li J, Dong Y, Zhu D. MMP-2 and MMP-9 contribute to the angiogenic effect produced by hypoxia/15-HETE in pulmonary endothelial cells. J Mol Cell Cardiol 2018; 121:36-50. [PMID: 29913136 DOI: 10.1016/j.yjmcc.2018.06.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 06/06/2018] [Accepted: 06/13/2018] [Indexed: 11/15/2022]
Abstract
Matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9) are the predominant gelatinases in the developing lung. Studies have shown that the expression of MMP-2 and MMP-9 is upregulated in hypoxic fibroblasts, 15-hydroxyeicosatetraenoic acid (15-HETE) regulated fibroblasts migration via modulating MMP-2 or MMP-9, and that hypoxia/15-HETE is a predominant contributor to the development of pulmonary arterial hypertension (PAH) through increased angiogenesis. However, the roles of MMP-2 and MMP-9 in pulmonary arterial endothelial cells (PAECs) angiogenesis as well as the molecular mechanism of hypoxia-regulated MMP-2 and MMP-9 expression have not been identified. The aim of this study was to investigate the role of MMP-2 and MMP-9 in PAEC proliferation and vascular angiogenesis and to determine the effects of hypoxia-induced 15-HETE on the expression of MMP-2 and MMP-9. Western blot, immunofluorescence, and real-time PCR were used to measure the expression of MMP-2 and MMP-9 in hypoxic PAECs. Immunohistochemical staining, flow cytometry, and tube formation as well as cell proliferation, viability, scratch-wound, and Boyden chamber migration assays were used to identify the roles and relationships between MMP-2, MMP-9, and 15-HETE in hypoxic PAECs. We found that hypoxia increased MMP-2 and MMP-9 expression in pulmonary artery endothelium both in vivo and in vitro in a time-dependent pattern. Moreover, administration of the MMP-2 and MMP-9 inhibitor MMI-166 significantly reversed hypoxia-induced increases in right ventricular systemic pressure (RVSP), right ventricular function, and thickening of the tunica media. Furthermore, up-regulation of MMP-2 and MMP-9 expression was induced by 15-HETE, which regulates PAEC proliferation, migration, and cell cycle transition that eventually leads to angiogenesis. Our study demonstrated that hypoxia increases the expression of MMP-2 and MMP-9 through the 15-lipoxygenase/15-HETE pathway, and that MMP-2 and MMP-9 promote PAEC angiogenesis. These findings suggest that MMP-2 and MMP-9 may serve as new potential therapeutic targets for the treatment of PAH.
Collapse
Affiliation(s)
- Ying Liu
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Hongyue Zhang
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Lixin Yan
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Wei Du
- School of Pharmacy, Harbin University of Commerce, Harbin, Heilongjiang Province, China
| | - Min Zhang
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - He Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lixin Zhang
- Department of Immunology, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, Heilongjiang Province, China
| | - Guangqun Li
- Medical Laboratory Technology, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Jijin Li
- Medical Laboratory Technology, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Yinchu Dong
- Medical Laboratory Technology, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Daling Zhu
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China.
| |
Collapse
|
11
|
Chan JKW, Bittner S, Bittner A, Atwal S, Shen WJ, Inayathullah M, Rajada J, Nicolls MR, Kraemer FB, Azhar S. Nordihydroguaiaretic Acid, a Lignan from Larrea tridentata (Creosote Bush), Protects Against American Lifestyle-Induced Obesity Syndrome Diet-Induced Metabolic Dysfunction in Mice. J Pharmacol Exp Ther 2018; 365:281-290. [PMID: 29472517 PMCID: PMC5878670 DOI: 10.1124/jpet.117.243733] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 02/16/2018] [Indexed: 12/30/2022] Open
Abstract
To determine the effects of nordihydroguaiaretic acid (NDGA) on metabolic and molecular changes in response to feeding a typical American fast food or Western diet, mice were fed an American lifestyle-induced obesity syndrome (ALIOS) diet and subjected to metabolic analysis. Male C57BL/6J mice were randomly assigned to the ALIOS diet, the ALIOS diet supplemented with NDGA (NDGA+ALIOS), or a control diet and were maintained on the specific diet for 8 weeks. Mice fed the ALIOS diet showed increased body, liver, and epididymal fat pad weight as well as increased plasma alanine transaminase (ALT) and aspartate aminotransferase (AST) levels (a measure of liver injury) and liver triglyceride content. Coadministration of NDGA normalized body and epididymal fat pad weight, ALT and AST levels, and liver triglycerides. NDGA treatment also improved insulin sensitivity but not glucose intolerance in mice fed the ALIOS diet. In mice fed the NDGA+ALIOS diet, NDGA supplementation induced peroxisome proliferator-activated receptor α (PPARα; the master regulator of fatty acid oxidation) and mRNA levels of carnitine palmitoyltransferases Cpt1c and Cpt2, key genes involved in fatty acid oxidation, compared with the ALIOS diet. NDGA significantly reduced liver endoplasmic reticulum (ER) stress response C/EBP homologous protein, compared with chow or the ALIOS diet, and also ameliorated ALIOS diet-induced elevation of apoptosis signaling protein, caspase 3. Likewise, NDGA downregulated the ALIOS diet-induced mRNA levels of Pparg, fatty acid synthase Fasn, and diacylglycerol acyltransferase Dgat2 NDGA treatment of ALIOS-fed mice upregulated the hepatic expression of antioxidant enzymes, glutathione peroxidase 4, and peroxiredoxin 3 proteins. In conclusion, we provide evidence that NDGA improves metabolic dysregulation by simultaneously modulating the PPARα transcription factor and key genes involved in fatty acid oxidation, key antioxidant and lipogenic enzymes, and apoptosis and ER stress signaling pathways.
Collapse
Affiliation(s)
- Jackie K W Chan
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.); and Division of Endocrinology, Gerontology, and Metabolism (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.), BioADD Laboratory, and Divisions of Cardiovascular Pharmacology CVI (M.I., J.R.) and Pulmonary and Critical Care Medicine (M.R.N.), Stanford University, Stanford, California
| | - Stefanie Bittner
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.); and Division of Endocrinology, Gerontology, and Metabolism (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.), BioADD Laboratory, and Divisions of Cardiovascular Pharmacology CVI (M.I., J.R.) and Pulmonary and Critical Care Medicine (M.R.N.), Stanford University, Stanford, California
| | - Alex Bittner
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.); and Division of Endocrinology, Gerontology, and Metabolism (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.), BioADD Laboratory, and Divisions of Cardiovascular Pharmacology CVI (M.I., J.R.) and Pulmonary and Critical Care Medicine (M.R.N.), Stanford University, Stanford, California
| | - Suman Atwal
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.); and Division of Endocrinology, Gerontology, and Metabolism (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.), BioADD Laboratory, and Divisions of Cardiovascular Pharmacology CVI (M.I., J.R.) and Pulmonary and Critical Care Medicine (M.R.N.), Stanford University, Stanford, California
| | - Wen-Jun Shen
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.); and Division of Endocrinology, Gerontology, and Metabolism (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.), BioADD Laboratory, and Divisions of Cardiovascular Pharmacology CVI (M.I., J.R.) and Pulmonary and Critical Care Medicine (M.R.N.), Stanford University, Stanford, California
| | - Mohammed Inayathullah
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.); and Division of Endocrinology, Gerontology, and Metabolism (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.), BioADD Laboratory, and Divisions of Cardiovascular Pharmacology CVI (M.I., J.R.) and Pulmonary and Critical Care Medicine (M.R.N.), Stanford University, Stanford, California
| | - Jayakumar Rajada
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.); and Division of Endocrinology, Gerontology, and Metabolism (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.), BioADD Laboratory, and Divisions of Cardiovascular Pharmacology CVI (M.I., J.R.) and Pulmonary and Critical Care Medicine (M.R.N.), Stanford University, Stanford, California
| | - Mark R Nicolls
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.); and Division of Endocrinology, Gerontology, and Metabolism (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.), BioADD Laboratory, and Divisions of Cardiovascular Pharmacology CVI (M.I., J.R.) and Pulmonary and Critical Care Medicine (M.R.N.), Stanford University, Stanford, California
| | - Fredric B Kraemer
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.); and Division of Endocrinology, Gerontology, and Metabolism (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.), BioADD Laboratory, and Divisions of Cardiovascular Pharmacology CVI (M.I., J.R.) and Pulmonary and Critical Care Medicine (M.R.N.), Stanford University, Stanford, California
| | - Salman Azhar
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.); and Division of Endocrinology, Gerontology, and Metabolism (J.K.W.C., S.B., A.B., S.At., W.-J.S., F.B.K., S.Az.), BioADD Laboratory, and Divisions of Cardiovascular Pharmacology CVI (M.I., J.R.) and Pulmonary and Critical Care Medicine (M.R.N.), Stanford University, Stanford, California
| |
Collapse
|
12
|
Qiao S, Mao G, Li H, Ma Z, Hong L, Zhang H, Wang C, An J. DPP-4 Inhibitor Sitagliptin Improves Cardiac Function and Glucose Homeostasis and Ameliorates β-Cell Dysfunction Together with Reducing S6K1 Activation and IRS-1 and IRS-2 Degradation in Obesity Female Mice. J Diabetes Res 2018; 2018:3641516. [PMID: 30116740 PMCID: PMC6079488 DOI: 10.1155/2018/3641516] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/08/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Chronic overnutrition leads to cardiac dysfunction and insulin (INS) resistance. Dipeptidyl peptidase-4 (DPP-4) improves glucose metabolism and insulin sensitivity in both human and animal models. In this study, we explored whether DPP-4 inhibitor sitagliptin (SIT) is involved in the protection of cardiac function and β-cell function using an obesity female mouse model. METHODS Six-week-old C57BL6/J mice were fed a high fat and fructose Western diet with DPP-4 inhibitor SIT for 12 weeks. Cardiac function was examined by echocardiography. Body weight, plasma glucose, and insulin concentrations were measured. The contents of total S6 kinase 1 (S6K1), phosphorylation of S6K1 activation, and INS docking proteins INS receptor substrates 1 and 2 (IRS-1, IRS-2) were assayed, and histology of heart tissue was performed. RESULTS Chronic Western diet consumption elevated plasma glucose and insulin and caused obesity, diastolic dysfunction, and β-cell dysfunction. DPP-4 inhibition with SIT resulted in reduction in body weight, fasting glucose, and plasma insulin, and improved cardiac diastolic dysfunction. SIT also decreased mTOR/S6K1 activation and prevented the degradation of IRS-1 and IRS-2. CONCLUSIONS This study revealed pleiotropic protective effects of DPP-4 inhibitor SIT on cardiac function, glycemia, and β-cell function together with reducing S6K1 activation and IRS-1 and IRS-2 degradation in the obesity female mouse model.
Collapse
Affiliation(s)
- Shigang Qiao
- Institute of Clinical Medicine Research, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
- Department of Pharmacology, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Soochow University, No. 199 Renai Road, Suzhou 215123, China
| | - Guofang Mao
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| | - Hua Li
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| | - Zhimin Ma
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| | - Lei Hong
- Institute of Clinical Medicine Research, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| | - Huiling Zhang
- Department of Pharmacology, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Soochow University, No. 199 Renai Road, Suzhou 215123, China
| | - Chen Wang
- Institute of Clinical Medicine Research, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| | - Jianzhong An
- Institute of Clinical Medicine Research, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| |
Collapse
|
13
|
Soto-Acosta R, Bautista-Carbajal P, Cervantes-Salazar M, Angel-Ambrocio AH, del Angel RM. DENV up-regulates the HMG-CoA reductase activity through the impairment of AMPK phosphorylation: A potential antiviral target. PLoS Pathog 2017; 13:e1006257. [PMID: 28384260 PMCID: PMC5383345 DOI: 10.1371/journal.ppat.1006257] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/24/2017] [Indexed: 01/01/2023] Open
Abstract
Dengue is the most common mosquito-borne viral disease in humans. Changes of lipid-related metabolites in endoplasmic reticulum of dengue virus (DENV) infected cells have been associated with replicative complexes formation. Previously, we reported that DENV infection inhibits HMGCR phosphorylation generating a cholesterol-enriched cellular environment in order to favor viral replication. In this work, using enzymatic assays, ELISA, and WB we found a significant higher activity of HMGCR in DENV infected cells, associated with the inactivation of AMPK. AMPK activation by metformin declined the HMGCR activity suggesting that AMPK inactivation mediates the enhanced activity of HMGCR. A reduction on AMPK phosphorylation activity was observed in DENV infected cells at 12 and 24 hpi. HMGCR and cholesterol co-localized with viral proteins NS3, NS4A and E, suggesting a role for HMGCR and AMPK activity in the formation of DENV replicative complexes. Furthermore, metformin and lovastatin (HMGCR inhibitor) altered this co-localization as well as replicative complexes formation supporting that active HMGCR is required for replicative complexes formation. In agreement, metformin prompted a significant dose-dependent antiviral effect in DENV infected cells, while compound C (AMPK inhibitor) augmented the viral genome copies and the percentage of infected cells. The PP2A activity, the main modulating phosphatase of HMGCR, was not affected by DENV infection. These data demonstrate that the elevated activity of HMGCR observed in DENV infected cells is mediated through AMPK inhibition and not by increase in PP2A activity. Interestingly, the inhibition of this phosphatase showed an antiviral effect in an HMGCR-independent manner. These results suggest that DENV infection increases HMGCR activity through AMPK inactivation leading to higher cholesterol levels in endoplasmic reticulum necessary for replicative complexes formation. This work provides new information about the mechanisms involved in host lipid metabolism during DENV replicative cycle and identifies new potential antiviral targets for DENV replication. DENV replicative complexes formation is associated with changes of lipid-related metabolites in endoplasmic reticulum, such as an increase in cholesterol synthesis. This increase correlates with a significant augment in the activity of HMGCoA reductase (the limiting enzyme in cholesterol synthesis), favoring a cholesterol-enriched cellular environment. The augment in the activity of the HMGCR observed in infected cells is caused by a decrease in the phosphorylation level of the HMGCR, associated with the inactivation of AMPK. In agreement, AMPK activation by metformin reduces HMGCR activity and affects viral replication. The role HMGCR and AMPK activity in DENV replicative complexes formation was confirmed by the co-localization of HMGCR and cholesterol with the viral proteins NS3, NS4A and E. Furthermore, metformin and lovastatin (HMGCR inhibitor) treatments altered this co-localization as well as replicative complexes formation supporting that active HMGCR is required for replicative complexes formation. The results show that during DENV infection, an increase in the HMGCR activity occurs through AMPK inactivation, leading to higher cholesterol levels in endoplasmic reticulum necessary for replicative complexes formation. This work provides new information about the mechanisms involved in host lipid metabolism during DENV replicative cycle and identifies potential new antiviral targets for DENV replication.
Collapse
Affiliation(s)
- Rubén Soto-Acosta
- Departmento de Infectómica y Patogénesis Molecular, CINVESTAV-IPN, México, D.F., México
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
| | | | | | | | - Rosa M. del Angel
- Departmento de Infectómica y Patogénesis Molecular, CINVESTAV-IPN, México, D.F., México
- * E-mail:
| |
Collapse
|
14
|
Dain A, Repossi G, Diaz-Gerevini GT, Vanamala J, Das UN, Eynard AR. Long chain polyunsaturated fatty acids (LCPUFAs) and nordihydroguaiaretic acid (NDGA) modulate metabolic and inflammatory markers in a spontaneous type 2 diabetes mellitus model (Stillman Salgado rats). Lipids Health Dis 2016; 15:205. [PMID: 27884155 PMCID: PMC5123226 DOI: 10.1186/s12944-016-0363-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 11/04/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a complex disease with alterations in metabolic and inflammatory markers. Stillman Salgado rats (eSS) spontaneously develop type 2 DM by middle age showing progressive impairment of glucose tolerance with hyperglycemia, hypertriglyceridemia and hyperinsulinemia. We analyzed the effects of supplementation of ω-3 and ω-6 polyunsaturated fatty acids (PUFAs) with or without nordihydroguaiaretic acid (NDGA) added, an antioxidant and lipoxygenase inhibitor, on metabolic and inflammatory parameters in eSS rats to evaluate whether they can delay development and/or prevent progression of DM. METHODS After weaning, eSS rats received, intraperitoneally, once a month ω-3 (EPA 35% and DHA 40%-6.25 mg/Kg) or ω-6 (90% arachidonic acid- 6. 25 mg/Kg) for twelve months. Two additional groups of rats received 1.9 mg/kg NDGA added to ω-3 and ω-6 fatty acids. Blood samples were collected at day 40, and at the end of the 6th month and 12th month of age to determine plasma triglycerides (TGs), total plasma fatty acids (FA), A1C hemoglobin (HbA1C), C-reactive protein (CRP), gamma glutamyl transpeptidase (GGT), lipo and hydro peroxides, nitrites and IL-6 (in plasma and liver, kidney, and pancreas) and underwent oral glucose tolerance test (OGTT) as well. Wistar and eSS rats that received saline solution were used as controls. RESULTS Plasma lipids profile, TG, fasting and post-prandial blood glucose levels, and glycosylated HbA1C showed significant improvements in ω-3 and ω-3 + NDGA treated animals compared to eSS control group. ω-3 and ω-3 + NDGA groups showed an inverse correlation with fasting blood glucose and showed lower plasma levels of GGT, TG, and CRP. eSS rats treated with ω-3 LCPUFAs showed reduced level of inflammatory and oxidative indices in plasma and liver, kidney and pancreas tissues in comparison with eSS control (non-treated) and ω-6 treated groups. CONCLUSIONS eSS rats are a useful model to study type 2 DM pathophysiology and related inflammatory indices. ω-3 + NDGA supplementation, at the doses tested, ameliorated inflammatory, metabolic and oxidative stress markers studied.
Collapse
Affiliation(s)
- Alejandro Dain
- Biología Celular, Histología y Embriología, Facultad de Ciencias Medicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| | - Gaston Repossi
- Biología Celular, Histología y Embriología, Facultad de Ciencias Medicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
- Cátedra de Histología, Embriología y Genética, Universidad Nacional de La Rioja, La Rioja, Argentina
- CONICET, Córdoba, Argentina
| | - Gustavo T Diaz-Gerevini
- Biología Celular, Histología y Embriología, Facultad de Ciencias Medicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| | - Jairam Vanamala
- Department of Food Science, Penn State University, 326 Food Science Building, University Park, PA, 16802, USA
| | - Undurti N Das
- UND Life Sciences, 2020 S 360th St, # K-202, Federal Way, WA, 98003, USA.
- BioScience Research Centre and Department of Medicine, GVP Hospital, Gayatri Vidya Parishad College of Engineering Campus, Visakhapatnam, 530 048, India.
| | - Aldo R Eynard
- Biología Celular, Histología y Embriología, Facultad de Ciencias Medicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina.
- CONICET, Córdoba, Argentina.
| |
Collapse
|
15
|
Zhang H, Shen WJ, Li Y, Bittner A, Bittner S, Tabassum J, Cortez YF, Kraemer FB, Azhar S. Microarray analysis of gene expression in liver, adipose tissue and skeletal muscle in response to chronic dietary administration of NDGA to high-fructose fed dyslipidemic rats. Nutr Metab (Lond) 2016; 13:63. [PMID: 27708683 PMCID: PMC5041401 DOI: 10.1186/s12986-016-0121-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 09/14/2016] [Indexed: 02/06/2023] Open
Abstract
Nordihydroguaiaretic acid (NDGA), the main metabolite of Creosote Bush, has been shown to have profound effects on the core components of metabolic syndrome, including lowering of blood glucose, free fatty acids and triglyceride levels, attenuating elevated blood pressure in several rodent models of dyslipidemia, and improving body weight, insulin resistance, diabetes and hypertension. In the present study, a high-fructose diet fed rat model of hypertriglyceridemia, dyslipidemia, insulin resistance and hepatic steatosis was employed to investigate the global transcriptional changes in the lipid metabolizing pathways in three insulin sensitive tissues: liver, skeletal muscle and adipose tissue in response to chronic dietary administration of NDGA. Sprague-Dawley male rats (SD) were fed a chow (control) diet, high-fructose diet (HFrD) or HFrD supplemented with NDGA (2.5 g/kg diet) for eight weeks. Dietary administration of NDGA decreased plasma levels of TG, glucose, and insulin, and attenuated hepatic TG accumulation. DNA microarray expression profiling indicated that dietary administration of NDGA upregulated the expression of certain genes involved in fatty acid oxidation and their transcription regulator, PPARα, decreased the expression of a number of lipogenic genes and relevant transcription factors, and differentially impacted the genes of fatty acid transporters, acetyl CoA synthetases, elongases, fatty acid desaturases and lipid clearance proteins in liver, skeletal muscle and adipose tissues. These findings suggest that NDGA ameliorates hypertriglyceridemia and steatosis primarily by inhibiting lipogenesis and enhancing fatty acid catabolism in three major insulin responsive tissues by altering the expression of key enzyme genes and transcription factors involved in de novo lipogenesis and fatty acid oxidation.
Collapse
Affiliation(s)
- Haiyan Zhang
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA USA ; Division of Endocrinology, Stanford University, Stanford, CA USA ; Present Address: Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Wen-Jun Shen
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA USA ; Division of Endocrinology, Stanford University, Stanford, CA USA
| | - Yihang Li
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA USA ; Division of Endocrinology, Stanford University, Stanford, CA USA ; Present Address: Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO USA
| | - Alex Bittner
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA USA
| | - Stefanie Bittner
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA USA
| | - Juveria Tabassum
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA USA
| | - Yuan F Cortez
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA USA
| | - Fredric B Kraemer
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA USA ; Division of Endocrinology, Stanford University, Stanford, CA USA
| | - Salman Azhar
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA USA ; Division of Endocrinology, Stanford University, Stanford, CA USA
| |
Collapse
|
16
|
Seong HA, Manoharan R, Ha H. Coordinate Activation of Redox-Dependent ASK1/TGF-β Signaling by a Multiprotein Complex (MPK38, ASK1, SMADs, ZPR9, and TRX) Improves Glucose and Lipid Metabolism in Mice. Antioxid Redox Signal 2016; 24:434-52. [PMID: 26421442 DOI: 10.1089/ars.2015.6325] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS To explore the molecular connections between redox-dependent apoptosis signal-regulating kinase 1 (ASK1) and transforming growth factor-β (TGF-β) signaling pathways and to examine the physiological processes in which coordinated regulation of these two signaling pathways plays a critical role. RESULTS We provide evidence that the ASK1 and TGF-β signaling pathways are interconnected by a multiprotein complex harboring murine protein serine-threonine kinase 38 (MPK38), ASK1, Sma- and Mad-related proteins (SMADs), zinc-finger-like protein 9 (ZPR9), and thioredoxin (TRX) and demonstrate that the activation of either ASK1 or TGF-β activity is sufficient to activate both the redox-dependent ASK1 and TGF-β signaling pathways. Physiologically, the restoration of the downregulated activation levels of ASK1 and TGF-β signaling in genetically and diet-induced obese mice by adenoviral delivery of SMAD3 or ZPR9 results in the amelioration of adiposity, hyperglycemia, hyperlipidemia, and impaired ketogenesis. INNOVATION AND CONCLUSION Our data suggest that the multiprotein complex linking ASK1 and TGF-β signaling pathways may be a potential target for redox-mediated metabolic complications.
Collapse
Affiliation(s)
- Hyun-A Seong
- Department of Biochemistry, School of Life Sciences, Chungbuk National University , Cheongju, Korea
| | - Ravi Manoharan
- Department of Biochemistry, School of Life Sciences, Chungbuk National University , Cheongju, Korea
| | - Hyunjung Ha
- Department of Biochemistry, School of Life Sciences, Chungbuk National University , Cheongju, Korea
| |
Collapse
|
17
|
Zhang H, Li Y, Hu J, Shen WJ, Singh M, Hou X, Bittner A, Bittner S, Cortez Y, Tabassum J, Kraemer FB, Azhar S. Effect of Creosote Bush-Derived NDGA on Expression of Genes Involved in Lipid Metabolism in Liver of High-Fructose Fed Rats: Relevance to NDGA Amelioration of Hypertriglyceridemia and Hepatic Steatosis. PLoS One 2015; 10:e0138203. [PMID: 26394137 PMCID: PMC4578927 DOI: 10.1371/journal.pone.0138203] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 08/27/2015] [Indexed: 12/11/2022] Open
Abstract
Nordihydroguaiaretic acid (NDGA), the main metabolite of Creosote bush, has been shown to have profound effects on the core components of the metabolic syndrome (MetS), lowering blood glucose, free fatty acids (FFA) and triglyceride (TG) levels in several models of dyslipidemia, as well as improving body weight (obesity), insulin resistance, diabetes and hypertension, and ameliorating hepatic steatosis. In the present study, a high-fructose diet (HFrD) fed rat model of hypertriglyceridemia was employed to further delineate the underlying mechanism by which NDGA exerts its anti-hypertriglyceridemic action. In the HFrD treatment group, NDGA administration by oral gavage decreased plasma levels of TG, glucose, FFA, and insulin, increased hepatic mitochondrial fatty acid oxidation and attenuated hepatic TG accumulation. qRT-PCR measurements indicated that NDGA treatment increased the mRNA expression of key fatty acid transport (L-FABP, CD36), and fatty acid oxidation (ACOX1, CPT-2, and PPARα transcription factor) genes and decreased the gene expression of enzymes involved in lipogenesis (FASN, ACC1, SCD1, L-PK and ChREBP and SREBP-1c transcription factors). Western blot analysis indicated that NDGA administration upregulated hepatic insulin signaling (P-Akt), AMPK activity (P-AMPK), MLYCD, and PPARα protein levels, but decreased SCD1, ACC1 and ACC2 protein content and also inactivated ACC1 activity (increased P-ACC1). These findings suggest that NDGA ameliorates hypertriglyceridemia and hepatic steatosis primarily by interfering with lipogenesis and promoting increased channeling of fatty acids towards their oxidation.
Collapse
Affiliation(s)
- Haiyan Zhang
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
- Division of Endocrinology, Stanford University, Stanford, California, United States of America
| | - Yihang Li
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
- Division of Endocrinology, Stanford University, Stanford, California, United States of America
| | - Jie Hu
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Wen-Jun Shen
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
- Division of Endocrinology, Stanford University, Stanford, California, United States of America
| | - Madhurima Singh
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
- Division of Endocrinology, Stanford University, Stanford, California, United States of America
| | - Xiaoming Hou
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
- Division of Endocrinology, Stanford University, Stanford, California, United States of America
| | - Alex Bittner
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Stefanie Bittner
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Yuan Cortez
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Juveria Tabassum
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Fredric B. Kraemer
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
- Division of Endocrinology, Stanford University, Stanford, California, United States of America
| | - Salman Azhar
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California, United States of America
- Division of Endocrinology, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
18
|
Hernández-Damián J, Andérica-Romero AC, Pedraza-Chaverri J. Paradoxical Cellular Effects and Biological Role of the Multifaceted Compound Nordihydroguaiaretic Acid. Arch Pharm (Weinheim) 2014; 347:685-97. [DOI: 10.1002/ardp.201400159] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 05/29/2014] [Accepted: 06/05/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Jacqueline Hernández-Damián
- Faculty of Chemistry, Department of Biology; National Autonomous University of Mexico (UNAM); University City D.F. Mexico
| | - Ana Cristina Andérica-Romero
- Faculty of Chemistry, Department of Biology; National Autonomous University of Mexico (UNAM); University City D.F. Mexico
| | - José Pedraza-Chaverri
- Faculty of Chemistry, Department of Biology; National Autonomous University of Mexico (UNAM); University City D.F. Mexico
| |
Collapse
|
19
|
Soto-Acosta R, Bautista-Carbajal P, Syed GH, Siddiqui A, Del Angel RM. Nordihydroguaiaretic acid (NDGA) inhibits replication and viral morphogenesis of dengue virus. Antiviral Res 2014; 109:132-40. [PMID: 25017471 DOI: 10.1016/j.antiviral.2014.07.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 07/03/2014] [Indexed: 12/22/2022]
Abstract
Dengue is the most common mosquito borne viral disease in humans. The infection with any of the 4 dengue virus serotypes (DENV) can either be asymptomatic or manifest in two clinical forms, the mild dengue fever or the more severe dengue hemorrhagic fever that may progress into dengue shock syndrome. A DENV replicative cycle relies on host lipid metabolism; specifically, DENV infection modulates cholesterol and fatty acid synthesis, generating a lipid-enriched cellular environment necessary for viral replication. Thus, the aim of this work was to evaluate the anti-DENV effect of the Nordihydroguaiaretic acid (NDGA), a hypolipidemic agent with antioxidant and anti-inflammatory properties. A dose-dependent inhibition in viral yield and NS1 secretion was observed in supernatants of infected cells treated for 24 and 48 h with different concentrations of NDGA. To evaluate the effect of NDGA in DENV replication, a DENV4 replicon transfected Vero cells were treated with different concentrations of NDGA. NDGA treatment significantly reduced DENV replication, reiterating the importance of lipids in viral replication. NDGA treatment also led to reduction in number of lipid droplets (LDs), the neutral lipid storage organelles involved in DENV morphogenesis that are known to increase in number during DENV infection. Furthermore, NDGA treatment resulted in dissociation of the C protein from LDs. Overall our results suggest that NDGA inhibits DENV infection by targeting genome replication and viral assembly.
Collapse
Affiliation(s)
- Rubén Soto-Acosta
- Departmento de Infectómica y Patogénesis molecular, CINVESTAV-IPN, México, D.F., Mexico
| | | | - Gulam H Syed
- Department of Medicine, Division of Infectious Disease, University of California, San Diego, La Jolla, CA, United States
| | - Aleem Siddiqui
- Department of Medicine, Division of Infectious Disease, University of California, San Diego, La Jolla, CA, United States
| | - Rosa M Del Angel
- Departmento de Infectómica y Patogénesis molecular, CINVESTAV-IPN, México, D.F., Mexico.
| |
Collapse
|
20
|
Kan CFK, Singh AB, Stafforini DM, Azhar S, Liu J. Arachidonic acid downregulates acyl-CoA synthetase 4 expression by promoting its ubiquitination and proteasomal degradation. J Lipid Res 2014; 55:1657-67. [PMID: 24879802 PMCID: PMC4109760 DOI: 10.1194/jlr.m045971] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Indexed: 01/16/2023] Open
Abstract
ACSL4 is a member of the long-chain acyl-CoA synthetase (ACSL) family with a marked preference for arachidonic acid (AA) as its substrate. Although an association between elevated levels of ACSL4 and hepatosteatosis has been reported, the function of ACSL4 in hepatic FA metabolism and the regulation of its functional expression in the liver remain poorly defined. Here we provide evidence that AA selectively downregulates ACSL4 protein expression in hepatic cells. AA treatment decreased the half-life of ACSL4 protein in HepG2 cells by approximately 4-fold (from 17.3 ± 1.8 h to 4.2 ± 0.4 h) without causing apoptosis. The inhibitory action of AA on ACSL4 protein stability could not be prevented by rosiglitazone or inhibitors that interfere with the cellular pathways involved in AA metabolism to biologically active compounds. In contrast, treatment of cells with inhibitors specific for the proteasomal degradation pathway largely prevented the AA-induced ACSL4 degradation. We further show that ACSL4 is intrinsically ubiquitinated and that AA treatment can enhance its ubiquitination. Collectively, our studies have identified a novel substrate-induced posttranslational regulatory mechanism by which AA downregulates ACSL4 protein expression in hepatic cells.
Collapse
Affiliation(s)
- Chin Fung Kelvin Kan
- Department of Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304
| | - Amar Bahadur Singh
- Department of Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304
| | | | - Salman Azhar
- Department of Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304
| | - Jingwen Liu
- Department of Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304
| |
Collapse
|
21
|
Harrison DE, Strong R, Allison DB, Ames BN, Astle CM, Atamna H, Fernandez E, Flurkey K, Javors MA, Nadon NL, Nelson JF, Pletcher S, Simpkins JW, Smith D, Wilkinson JE, Miller RA. Acarbose, 17-α-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males. Aging Cell 2014; 13:273-82. [PMID: 24245565 PMCID: PMC3954939 DOI: 10.1111/acel.12170] [Citation(s) in RCA: 323] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2013] [Indexed: 01/09/2023] Open
Abstract
Four agents — acarbose (ACA), 17-α-estradiol (EST), nordihydroguaiaretic acid (NDGA), and methylene blue (MB) — were evaluated for lifespan effects in genetically heterogeneous mice tested at three sites. Acarbose increased male median lifespan by 22% (P < 0.0001), but increased female median lifespan by only 5% (P = 0.01). This sexual dimorphism in ACA lifespan effect could not be explained by differences in effects on weight. Maximum lifespan (90th percentile) increased 11% (P < 0.001) in males and 9% (P = 0.001) in females. EST increased male median lifespan by 12% (P = 0.002), but did not lead to a significant effect on maximum lifespan. The benefits of EST were much stronger at one test site than at the other two and were not explained by effects on body weight. EST did not alter female lifespan. NDGA increased male median lifespan by 8–10% at three different doses, with P-values ranging from 0.04 to 0.005. Females did not show a lifespan benefit from NDGA, even at a dose that produced blood levels similar to those in males, which did show a strong lifespan benefit. MB did not alter median lifespan of males or females, but did produce a small, statistically significant (6%, P = 0.004) increase in female maximum lifespan. These results provide new pharmacological models for exploring processes that regulate the timing of aging and late-life diseases, and in particular for testing hypotheses about sexual dimorphism in aging and health.
Collapse
Affiliation(s)
| | - Randy Strong
- Barshop Institute for Longevity and Aging Studies The University of Texas Health Science Center at San Antonio San Antonio TX 78245USA
- Geriatric Research, Education and Clinical Center South Texas Veterans Health Care System San Antonio TX 78229USA
- Research Service South Texas Veterans Health Care System San Antonio TX 78229USA
- Department of Pharmacology The University of Texas Health Science Center at San Antonio San Antonio TX 78229USA
| | - David B. Allison
- Department of Biostatistics University of Alabama at Birmingham Birmingham AL 35294USA
| | - Bruce N. Ames
- Children's Hospital Oakland Research Institute 5700 Martin Luther King Jr. Way Oakland CA 94609‐1673USA
| | | | - Hani Atamna
- Children's Hospital Oakland Research Institute 5700 Martin Luther King Jr. Way Oakland CA 94609‐1673USA
| | - Elizabeth Fernandez
- Barshop Institute for Longevity and Aging Studies The University of Texas Health Science Center at San Antonio San Antonio TX 78245USA
- Geriatric Research, Education and Clinical Center South Texas Veterans Health Care System San Antonio TX 78229USA
- Research Service South Texas Veterans Health Care System San Antonio TX 78229USA
- Department of Pharmacology The University of Texas Health Science Center at San Antonio San Antonio TX 78229USA
| | | | - Martin A. Javors
- Barshop Institute for Longevity and Aging Studies The University of Texas Health Science Center at San Antonio San Antonio TX 78245USA
- Department of Psychiatry The University of Texas Health Science Center at San Antonio San Antonio TX 78229USA
| | - Nancy L. Nadon
- Division of Aging Biology National Institute on Aging Bethesda MD 20892USA
| | - James F. Nelson
- Barshop Institute for Longevity and Aging Studies The University of Texas Health Science Center at San Antonio San Antonio TX 78245USA
- Department of Physiology The University of Texas Health Science Center at San Antonio San Antonio TX 78229USA
| | - Scott Pletcher
- Department of Molecular and Integrative Physiology, and Geriatrics Center University of Michigan Ann Arbor MI 48109USA
| | - James W. Simpkins
- Department of Pharmacology & Neuroscience University of North Texas Health Science Center Fort Worth TX 76107USA
| | - Daniel Smith
- Department of Nutrition Sciences University of Alabama at Birmingham Birmingham AL 35294USA
| | - J. Erby Wilkinson
- Unit for Laboratory Animal Medicine University of Michigan School of Medicine Ann Arbor MI 48109USA
| | - Richard A. Miller
- Department of Pathology and Geriatrics Center University of Michigan Ann Arbor MI 48109USA
| |
Collapse
|
22
|
Ferreira AVM, Menezes-Garcia Z, Mario EG, Delpuerto HL, Martins AS, Botion LM. Increased expression of oxidative enzymes in adipose tissue following PPARα-activation. Metabolism 2014; 63:456-60. [PMID: 24439670 DOI: 10.1016/j.metabol.2013.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 12/05/2013] [Accepted: 12/17/2013] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Evaluate the effect of fenofibrate treatment on the expression of PPARα and oxidative enzymes in adipose tissue. MATERIALS/METHODS Wistar male rats were fed a balanced diet supplemented with 100mg.Kg-1 bw.day-1 fenofibrate (Sigma) during nine days. Plasma glucose, free fatty acids (FFA) leptin and insulin were determined. PPARα, ACO and CPT-1 mRNA expression and amount of PPARα and PPARγ protein were assessed in epididymal adipose tissue. Oral glucose tolerance test was evaluated into overnight fasted rats. Glucose uptake was measured in adipocytes isolated from epididymal fat pads in the presence or absence of insulin (25ng/mL). RESULTS Fenofibrate treatment increased PPARα and PPARγ protein abundance in adipose tissue. In addition to it well- known effect on oxidative enzymes in liver, fenofibrate treatment also induces a high expression of Acyl CoA Oxidase (ACO) and Carnitine palmitoyltransferase 1 (CPT-1) in adipose tissue. Furthermore, we have shown that the fenofibrate treatment improves the glucose tolerance and enhance the glucose uptake by adipocytes. CONCLUSION Altogether, the data suggest that fenofibrate have a direct effect in adipose tissue contributing to the low adiposity and improvement of glucose homeostasis.
Collapse
Affiliation(s)
| | - Zélia Menezes-Garcia
- Department of Microbiology, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Erica Guilhen Mario
- Department of Physiology and Biophysics, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Helen Lima Delpuerto
- Department of Physiology and Biophysics, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Almir Souza Martins
- Department of Physiology and Biophysics, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Leida Maria Botion
- Department of Physiology and Biophysics, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|