1
|
Lupu A, Stoleriu G, Nedelcu AH, Perju SN, Gavrilovici C, Baciu G, Mihai CM, Chisnoiu T, Morariu ID, Grigore E, Shawais SK, Salaru DL, Revenco N, Lupu VV. Overview of Oxidative Stress in Systemic Lupus Erythematosus. Antioxidants (Basel) 2025; 14:303. [PMID: 40227251 PMCID: PMC11939823 DOI: 10.3390/antiox14030303] [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: 01/08/2025] [Revised: 02/16/2025] [Accepted: 02/27/2025] [Indexed: 04/15/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that is frequently diagnosed in female patients, caused by multiple interacting factors. It has a complex pathogenesis which can affect almost any organ, from the kidneys to the cardiovascular, pulmonary, neurological, osteoarticular, and hematological systems. The present narrative review seeks to elucidate the role of reactive oxygen species (ROS) in the pathogenesis of SLE. The central question guiding this study is to what extent these serum protein modifications correlate with disease activity and organ damage in SLE. It is characterized by the decreased apoptosis and increased necrosis of T cells and the NETosis of granulocytes. Given the impact of an SLE diagnosis on one's life, this narrative review aims to evaluate the intricacies of oxidative stress and its relevance to the pathogenesis and treatment of the disease. Topics such as understanding processes of oxidative stress, their damaging pathways, oxidative stress biomarkers, and their role in the future assistance of clinical decisions will be discussed in the article. The accurate determination of biomarkers is taught to improve both the diagnosis and the management of the disease, while antioxidant therapy may open a new door for the treatment.
Collapse
Affiliation(s)
- Ancuta Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (C.G.); (V.V.L.)
| | - Gabriela Stoleriu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.S.); (G.B.)
| | - Alin Horatiu Nedelcu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (S.K.S.); (D.L.S.)
| | - Sara Nadeea Perju
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (C.G.); (V.V.L.)
| | - Cristina Gavrilovici
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (C.G.); (V.V.L.)
| | - Ginel Baciu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.S.); (G.B.)
| | - Cristina Maria Mihai
- Pediatrics, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania; (C.M.M.); (T.C.)
| | - Tatiana Chisnoiu
- Pediatrics, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania; (C.M.M.); (T.C.)
| | - Ionela Daniela Morariu
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Ecaterina Grigore
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (S.K.S.); (D.L.S.)
| | - Shwan Karwan Shawais
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (S.K.S.); (D.L.S.)
| | - Delia Lidia Salaru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (S.K.S.); (D.L.S.)
| | - Ninel Revenco
- Pediatrics, “Nicolae Testemitanu” State University of Medicine and Pharmacy, 2004 Chisinau, Moldova;
| | - Vasile Valeriu Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (C.G.); (V.V.L.)
| |
Collapse
|
2
|
Petrella L, Polito R, Catapano A, Santillo A, Ciliberti MG, Sevi A, Messina A, Cavaliere G, Marino F, Polverino MG, Messina G, Monda M, Mollica MP, Crispino M, Cimmino F, Albenzio M, Trinchese G. Goat Milk Supplementation Modulates the Mitochondrial Metabolic Flexibility and Orexin-A Levels Influencing the Inflammatory Pattern in Rats. Antioxidants (Basel) 2024; 13:1054. [PMID: 39334713 PMCID: PMC11429022 DOI: 10.3390/antiox13091054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Milk and its derivatives are included in a balanced diet of humans as excellent sources of proteins, vitamins, and essential minerals that are functional nutrients. Knowledge about the nutritional benefits or harms due to milk consumption has been expanding in recent years. We previously explored, in rodent models, the metabolic effects of isoenergetic intake of milk derived from cows, donkeys, or humans, while the impact of goat's milk intake has remained unexplored. The aim of this work was to investigate, in an animal model, the effects of dietary supplementation with goat's milk on energy homeostasis and inflammatory state, focusing on the modulation of mitochondrial functions in most metabolically active organs, such as skeletal muscle and the liver. In addition, we highlighted a link between nutrient intake, substrate metabolism, and the orexinergic system. Our results indicate that goat milk improves mitochondrial oxidative capacity and reduces inflammation and oxidative stress in both organs. Notably, goat milk lowers the circulating levels of Orexin-A, a neuropeptide that plays a crucial role in regulating peripheral energy balance and central nervous system mechanisms. These data provide the first evidence that the anti-inflammatory and antioxidant effects of goat milk are mediated by the modulation of mitochondrial functions and orexinergic signaling.
Collapse
Affiliation(s)
- Lidia Petrella
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Rita Polito
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy
| | - Angela Catapano
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Antonella Santillo
- Department of Agriculture, Food, Natural Resources and Engineering (DAFNE), University of Foggia, 71100 Foggia, Italy
| | - Maria Giovanna Ciliberti
- Department of Agriculture, Food, Natural Resources and Engineering (DAFNE), University of Foggia, 71100 Foggia, Italy
| | - Agostino Sevi
- Department of Agriculture, Food, Natural Resources and Engineering (DAFNE), University of Foggia, 71100 Foggia, Italy
| | - Antonietta Messina
- Department of Precision Medicine, University of Campania, Luigi Vanvitelli, 80131 Naples, Italy
| | - Gina Cavaliere
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| | - Francesca Marino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | | | - Giovanni Messina
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Marzia Albenzio
- Department of Agriculture, Food, Natural Resources and Engineering (DAFNE), University of Foggia, 71100 Foggia, Italy
| | - Giovanna Trinchese
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| |
Collapse
|
3
|
Melini S, Trinchese G, Lama A, Cimmino F, Del Piano F, Comella F, Opallo N, Leo A, Citraro R, Trabace L, Mattace Raso G, Pirozzi C, Mollica MP, Meli R. Sex Differences in Hepatic Inflammation, Lipid Metabolism, and Mitochondrial Function Following Early Lipopolysaccharide Exposure in Epileptic WAG/Rij Rats. Antioxidants (Basel) 2024; 13:957. [PMID: 39199203 PMCID: PMC11351225 DOI: 10.3390/antiox13080957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
Among the non-communicable neurological diseases, epilepsy is characterized by abnormal brain activity with several peripheral implications. The role of peripheral inflammation in the relationship between seizure development and nonalcoholic fatty liver disease based on sex difference remains still overlooked. Severe early-life infections lead to increased inflammation that can aggravate epilepsy and hepatic damage progression, both related to increased odds of hospitalization for epileptic patients with liver diseases. Here, we induced a post-natal-day 3 (PND3) infection by LPS (1 mg/kg, i.p.) to determine the hepatic damage in a genetic model of young epileptic WAG/Rij rats (PND45). We evaluated intra- and inter-gender differences in systemic and liver inflammation, hepatic lipid dysmetabolism, and oxidative damage related to mitochondrial functional impairment. First, epileptic rats exposed to LPS, regardless of gender, displayed increased serum hepatic enzymes and altered lipid profile. Endotoxin challenge triggered a more severe inflammatory and immune response in male epileptic rats, compared to females in both serum and liver, increasing pro-inflammatory cytokines and hepatic immune cell recruitment. Conversely, LPS-treated female rats showed significant alterations in systemic and hepatic lipid profiles and reduced mitochondrial fatty acid oxidation. The two different sex-dependent mechanisms of LPS-induced liver injury converge in increased ROS production and related mitochondrial oxidative damage in both sexes. Notably, a compensatory increase in antioxidant defense was evidenced only in female rats. Our study with a translational potential demonstrates, for the first time, that early post-natal infections in epileptic rats induced or worsened hepatic disorders in a sex-dependent manner, amplifying inflammation, lipid dysmetabolism, and mitochondrial impairment.
Collapse
Affiliation(s)
- Stefania Melini
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (S.M.); (A.L.); (F.C.); (N.O.); (G.M.R.); (R.M.)
| | - Giovanna Trinchese
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.T.); (F.C.); (M.P.M.)
| | - Adriano Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (S.M.); (A.L.); (F.C.); (N.O.); (G.M.R.); (R.M.)
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.T.); (F.C.); (M.P.M.)
| | - Filomena Del Piano
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy;
| | - Federica Comella
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (S.M.); (A.L.); (F.C.); (N.O.); (G.M.R.); (R.M.)
| | - Nicola Opallo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (S.M.); (A.L.); (F.C.); (N.O.); (G.M.R.); (R.M.)
| | - Antonio Leo
- Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (A.L.); (R.C.)
| | - Rita Citraro
- Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (A.L.); (R.C.)
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Giuseppina Mattace Raso
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (S.M.); (A.L.); (F.C.); (N.O.); (G.M.R.); (R.M.)
| | - Claudio Pirozzi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (S.M.); (A.L.); (F.C.); (N.O.); (G.M.R.); (R.M.)
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.T.); (F.C.); (M.P.M.)
| | - Rosaria Meli
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (S.M.); (A.L.); (F.C.); (N.O.); (G.M.R.); (R.M.)
| |
Collapse
|
4
|
Cuciniello R, Luongo D, Maurano F, Crispi S, Bergamo P. Dietary conjugated linoleic acid downregulates the AlCl 3-induced hyperactivation of compensatory and maladaptive signalling in the mouse brain cortex. Free Radic Biol Med 2024; 213:102-112. [PMID: 38218550 DOI: 10.1016/j.freeradbiomed.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/23/2023] [Accepted: 01/04/2024] [Indexed: 01/15/2024]
Abstract
Oxidative stress, hyperactivation of compensatory mechanisms (unfolded protein response, UPR; nuclear factor erythroid 2-related factor 2, Nrf2) and the stimulation of maladaptive response (inflammation/apoptosis) are interconnected pathogenic processes occurring during Alzheimer's disease (AD) progression. The neuroprotective ability of dietary Conjugated linoleic acid (CLAmix) in a mouse model of AlCl3-induced AD was recently described but, the effects of AlCl3 or CLAmix intake on these pathogenic processes are still unknown. The effects of dietary AlCl3 or CLAmix - alone and in combination - were examined in the brain cortex of twenty-eight BalbC mice divided into 4 groups (n = 7 each). The neurotoxic effects of AlCl3 were investigated in animals treated for 5 weeks with 100 mg/kg/day (AL). CLAmix supplementation (600 mg/kg bw/day) for 7 weeks (CLA) was aimed at evaluating its modulatory effects on the Nrf2 pathway while its co-treatment with AlCl3 during the last 5 weeks of CLAmix intake (CLA + AL) was used to investigate its neuroprotective ability. Untreated mice were used as controls. In the CLA group, the NADPH oxidase (NOX) activation in the brain cortex was accompanied by the modulation of the Nrf2 pathway. By contrast, in the AL mice, the significant upregulation of oxidative stress markers, compensatory pathways (UPR/Nrf2), proinflammatory cytokines (IL-6, TNFα) and the proapoptotic protein Bax levels were found as compared with control. Notably, in CLA + AL mice, the marked decrease of oxidative stress, UPR/Nrf2 markers and proinflammatory cytokines levels were associated with the significant increase of the antiapoptotic protein Bcl2. The involvement of NOX in the adaptive response elicited by CLAmix along with its protective effects against the onset of several pathogenic processes triggered by AlCl3, broadens the knowledge of the mechanism underlying the pleiotropic activity of Nrf2 activators and sheds new light on their potential therapeutic use against neurodegenerative disorders.
Collapse
Affiliation(s)
- R Cuciniello
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), Naples, 80100, Italy; IRCCS Neuromed, Pozzilli, 86077, Isernia, Italy
| | - D Luongo
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, 83100, Italy
| | - F Maurano
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, 83100, Italy
| | - S Crispi
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), Naples, 80100, Italy
| | - P Bergamo
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), Naples, 80100, Italy.
| |
Collapse
|
5
|
Yan Z, Chen Q, Xia Y. Oxidative Stress Contributes to Inflammatory and Cellular Damage in Systemic Lupus Erythematosus: Cellular Markers and Molecular Mechanism. J Inflamm Res 2023; 16:453-465. [PMID: 36761905 PMCID: PMC9907008 DOI: 10.2147/jir.s399284] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease with complex pathogenesis, the treatment of which relies exclusively on the use of immunosuppressants. Increased oxidative stress is involved in causing inflammatory and cellular defects in the pathogenesis of SLE. Various inflammatory and cellular markers including oxidative modifications of proteins, lipids, and DNA contribute to immune system dysregulation and trigger an aggressive autoimmune attack through molecular mechanisms like enhanced NETosis, mTOR pathway activation, and imbalanced T-cell differentiation. Accordingly, the detection of inflammatory and cellular markers is important for providing an accurate assessment of the extent of oxidative stress. Oxidative stress also reduces DNA methylation, thus allowing the increased expression of affected genes. As a result, pharmacological approaches targeting oxidative stress yield promising results in treating patients with SLE. The purpose of this review is to examine the involvement of oxidative stress in the pathogenesis and management of SLE.
Collapse
Affiliation(s)
- Zhu Yan
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Qin Chen
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China,Correspondence: Yumin Xia, Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, 157 Xiwu Road, Xi’an, 710004, People’s Republic of China, Tel/Fax +86-29-87679969, Email
| |
Collapse
|
6
|
He J, Ma C, Tang D, Zhong S, Yuan X, Zheng F, Zeng Z, Chen Y, Liu D, Hong X, Dai W, Yin L, Dai Y. Absolute quantification and characterization of oxylipins in lupus nephritis and systemic lupus erythematosus. Front Immunol 2022; 13:964901. [PMID: 36275708 PMCID: PMC9582137 DOI: 10.3389/fimmu.2022.964901] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/21/2022] [Indexed: 12/02/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with multi-organ inflammation and defect, which is linked to many molecule mediators. Oxylipins as a class of lipid mediator have not been broadly investigated in SLE. Here, we applied targeted mass spectrometry analysis to screen the alteration of oxylipins in serum of 98 SLE patients and 106 healthy controls. The correlation of oxylipins to lupus nephritis (LN) and SLE disease activity, and the biomarkers for SLE classification, were analyzed. Among 128 oxylipins analyzed, 92 were absolutely quantified and 26 were significantly changed. They were mainly generated from the metabolism of several polyunsaturated fatty acids, including arachidonic acid (AA), linoleic acid (LA), docosahexanoic acid (DHA), eicosapentanoic acid (EPA) and dihomo-γ-linolenic acid (DGLA). Several oxylipins, especially those produced from AA, showed different abundance between patients with and without lupus nephritis (LN). The DGLA metabolic activity and DGLA generated PGE1, were significantly associated with SLE disease activity. Random forest-based machine learning identified a 5-oxylipin combination as potential biomarker for SLE classification with high accuracy. Seven individual oxylipin biomarkers were also identified with good performance in distinguishing SLE patients from healthy controls (individual AUC > 0.7). Interestingly, the biomarkers for differentiating SLE patients from healthy controls are distinct from the oxylipins differentially expressed in LN patients vs. non-LN patients. This study provides possibilities for the understanding of SLE characteristics and the development of new tools for SLE classification.
Collapse
Affiliation(s)
- Jingquan He
- Department of Radiotherapy, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen, China
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Chiyu Ma
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Donge Tang
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Shaoyun Zhong
- Biotree Metabolomics Research Center, Biotree, Shanghai, China
| | - Xiaofang Yuan
- Biotree Metabolomics Research Center, Biotree, Shanghai, China
| | - Fengping Zheng
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Zhipeng Zeng
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Yumei Chen
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Dongzhou Liu
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Xiaoping Hong
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Weier Dai
- College of Natural Science, University of Texas at Austin, Austin, TX, United States
| | - Lianghong Yin
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yong Dai
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
- *Correspondence: Yong Dai,
| |
Collapse
|
7
|
Cuciniello R, Luongo D, Ferramosca A, Lunetti P, Rotondi-Aufiero V, Crispi S, Zara V, Maurano F, Filosa S, Bergamo P. Conjugated linoleic acid downregulates Alzheimer's hallmarks in aluminum mouse model through an Nrf2-mediated adaptive response and increases brain glucose transporter levels. Free Radic Biol Med 2022; 191:48-58. [PMID: 36028179 DOI: 10.1016/j.freeradbiomed.2022.08.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/15/2022] [Indexed: 10/15/2022]
Abstract
Mitochondrial dysfunction, oxidative stress, inflammation and glucose dysmetabolism are pathological signs of Alzheimer's disease (AD). Dietary aluminum (Al) overload is often used to induce AD in rodents and trigger the onset of oxidative-stress hallmarks resembling those of the human disease. The Nuclear factor erythroid 2-related factor 2 (Nrf2), owing to its key role in redox homeostasis, mitochondrial function and inflammation, is a promising drug target for neurological disorders, but only a few data are available on its modulatory effects on glucose transporter expression levels. While it has been found that the protective effect of Conjugated linoleic acid (CLA) occurs through the activation of an Nrf2-mediated adaptive response, its beneficial effect on the considered pathological signs in the Al-induced model has not been established yet. Thirty-five male BalbC mice were divided into 5 groups: two Al-intoxicated groups were treated for 5 weeks with low or high Al doses (8 or 100 mg/kg/day in drinking water, respectively; L or H). Two groups of animals, orally supplemented with CLA (600 mg/kg bw/day) for 7 weeks (2 preliminary weeks plus the 5-week treatment with Al; CLA + L, CLA + H) were used to investigate its protective effect, while untreated mice were used as control (Cntr). We provide evidence that mitochondrial dysfunction, Nrf2 alteration, inflammation and Acetylcholinesterase (AChE) hyperactivation can occur even from L exposure. Interestingly, animal pre-treatment with an allometric CLA dose led to significant downregulation of the toxic effects elicited by L or H, likely through the activation of an adaptive response. In conclusion, CLA ability to increase the level of glucose transporters - along with its antioxidant and anti-inflammatory effect - expands the therapeutic targets of these molecules and comes out as an intriguing suitable candidate for the treatment of multifactorial disease.
Collapse
Affiliation(s)
- R Cuciniello
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), 80100, Naples, Italy; IRCCS Neuromed, 86077, Pozzilli, IS, Italy
| | - D Luongo
- Institute of Food Sciences, National Research Council (CNR-ISA), 83100, Avellino, Italy
| | - A Ferramosca
- Department of Environmental and Biological Sciences and Technologies, University of Salento, 73100, Lecce, Italy
| | - P Lunetti
- Department of Environmental and Biological Sciences and Technologies, University of Salento, 73100, Lecce, Italy
| | - V Rotondi-Aufiero
- Institute of Food Sciences, National Research Council (CNR-ISA), 83100, Avellino, Italy
| | - S Crispi
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), 80100, Naples, Italy
| | - V Zara
- Department of Environmental and Biological Sciences and Technologies, University of Salento, 73100, Lecce, Italy
| | - F Maurano
- Institute of Food Sciences, National Research Council (CNR-ISA), 83100, Avellino, Italy
| | - S Filosa
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), 80100, Naples, Italy; IRCCS Neuromed, 86077, Pozzilli, IS, Italy
| | - P Bergamo
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), 80100, Naples, Italy.
| |
Collapse
|
8
|
Kaura S, Parle M, Insa R, Yadav BS, Sethi N. Neuroprotective effect of goat milk. Small Rumin Res 2022. [DOI: 10.1016/j.smallrumres.2022.106748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
9
|
Chen Q, Wang J, Xiang M, Wang Y, Zhang Z, Liang J, Xu J. The Potential Role of Ferroptosis in Systemic Lupus Erythematosus. Front Immunol 2022; 13:855622. [PMID: 35529869 PMCID: PMC9068945 DOI: 10.3389/fimmu.2022.855622] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that is accompanied with autoantibody production and inflammation. Other features of SLE pathogenesis include iron accumulation, oxidative stress, and lipid peroxidation, which are also major biochemical characteristics of ferroptosis, a novel non-apoptotic regulated form of cell death. To date, ferroptosis has been demonstrated to be an important driver of lupus progression, and several ferroptosis inhibitors have therapeutic effect in lupus-prone mice. Given the emerging link between ferroptosis and SLE, it can be postulated that ferroptosis is an integral component in the vicious cycle of immune dysfunction, inflammation, and tissue damage in SLE pathogenesis. In this review, we summarize the potential links between ferroptosis and SLE, with the aim of elucidating the underlying pathogenic mechanism of ferroptosis in lupus, and providing a new promising therapeutic strategy for SLE.
Collapse
Affiliation(s)
| | | | | | | | | | - Jun Liang
- *Correspondence: Jun Liang, ; Jinhua Xu,
| | - Jinhua Xu
- *Correspondence: Jun Liang, ; Jinhua Xu,
| |
Collapse
|
10
|
Trinchese G, Cimmino F, Cavaliere G, Rosati L, Catapano A, Sorriento D, Murru E, Bernardo L, Pagani L, Bergamo P, Scudiero R, Iaccarino G, Greco L, Banni S, Crispino M, Mollica MP. Heart Mitochondrial Metabolic Flexibility and Redox Status Are Improved by Donkey and Human Milk Intake. Antioxidants (Basel) 2021; 10:antiox10111807. [PMID: 34829678 PMCID: PMC8614950 DOI: 10.3390/antiox10111807] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/03/2021] [Accepted: 11/11/2021] [Indexed: 01/24/2023] Open
Abstract
The biological mechanisms linking nutrition and antioxidants content of the diet with cardiovascular protection are subject of intense investigation. It has been demonstrated that dietary supplementation with cow, donkey or human milk, characterized by distinct nutritional properties, triggers significant differences in the metabolic and inflammatory status through the modulation of hepatic and skeletal muscle mitochondrial functions. Cardiac mitochondria play a key role for energy-demanding heart functions, and their disfunctions is leading to pathologies. Indeed, an altered heart mitochondrial function and the consequent increased reactive oxygen species (ROS) production and inflammatory state, is linked to several cardiac diseases such as hypertension and heart failure. In this work it was investigated the impact of the milk consumption on heart mitochondrial functions, inflammation and oxidative stress. In addition, it was underlined the crosstalk between mitochondrial metabolic flexibility, lipid storage and redox status as control mechanisms for the maintenance of cardiovascular health.
Collapse
Affiliation(s)
- Giovanna Trinchese
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.T.); (F.C.); (G.C.); (L.R.); (A.C.); (R.S.); (M.C.)
- BAT Centre—Interuniversity Centre for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Naples, Italy
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.T.); (F.C.); (G.C.); (L.R.); (A.C.); (R.S.); (M.C.)
| | - Gina Cavaliere
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.T.); (F.C.); (G.C.); (L.R.); (A.C.); (R.S.); (M.C.)
| | - Luigi Rosati
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.T.); (F.C.); (G.C.); (L.R.); (A.C.); (R.S.); (M.C.)
- BAT Centre—Interuniversity Centre for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Naples, Italy
| | - Angela Catapano
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.T.); (F.C.); (G.C.); (L.R.); (A.C.); (R.S.); (M.C.)
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy; (D.S.); (G.I.)
| | - Elisabetta Murru
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (E.M.); (S.B.)
| | - Luca Bernardo
- Department of Childhood and Developmental Medicine, ASST Fatebenefratelli-Sacco, 20157 Milan, Italy; (L.B.); (L.P.)
| | - Luciana Pagani
- Department of Childhood and Developmental Medicine, ASST Fatebenefratelli-Sacco, 20157 Milan, Italy; (L.B.); (L.P.)
| | - Paolo Bergamo
- Institute of Bioscience and Bioresources CNR, IBBR-UOS, 80131 Naples, Italy;
| | - Rosaria Scudiero
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.T.); (F.C.); (G.C.); (L.R.); (A.C.); (R.S.); (M.C.)
- BAT Centre—Interuniversity Centre for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Naples, Italy
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy; (D.S.); (G.I.)
| | - Luigi Greco
- Department of Translational Medical Sciences, Section of Pediatrics, University of Naples Federico II, 80131 Naples, Italy;
| | - Sebastiano Banni
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (E.M.); (S.B.)
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.T.); (F.C.); (G.C.); (L.R.); (A.C.); (R.S.); (M.C.)
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (G.T.); (F.C.); (G.C.); (L.R.); (A.C.); (R.S.); (M.C.)
- BAT Centre—Interuniversity Centre for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80100 Naples, Italy
- Correspondence: ; Tel.: +39-081-679-990
| |
Collapse
|
11
|
Aydın B, Güler Şahin C, Şekeroğlu V, Atlı Şekeroğlu Z. Conjugated linoleic acid protects brain mitochondrial function in acrolein induced male rats. Toxicol Mech Methods 2021; 31:674-679. [PMID: 34238125 DOI: 10.1080/15376516.2021.1952673] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Acrolein (AC) is a toxic substance that can have a neurotoxic effect. It can cause oxidative stress and mitochondrial dysfunction. Conjugated linoleic acid (CLA), a dietary supplement, has many biological functions. Limited information is available about the effect of CLA on AC-induced brain toxicity. Therefore, the present study aims to investigate the effect of CLA on mitochondrial oxidative stress, respiratory enzymes, krebs cycle enzymes and ATP levels in AC treated rat brain. Sprague Dawley male rats were given AC (5 mg/kg i.p.), CLA (200 mg/kg orally) and CLA with AC for six days per week for 30 days. Some oxidative stress parameters and mitochondrial enzymes such as manganese super oxide dismutase, glutathione peroxidase, NADP+-dependent isocitrate dehydrogenase (ICDH), alpha-ketoglutarate dehydrogenase (α-KGDH), malate dehydrogenase, reduced glutathione (GSH), lipid peroxidation (LP), protein carbonyl (PC), oxidative phosphorylation (OXPHOS) and tricarboxylic acid cycle (TCA) enzymes, and ATP levels were determined. AC significantly decreased the activities of GSH, antioxidant enzymes, OXPHOS enzymes (complex I and IV), TCA enzymes (ICDH and α-KGDH) and ATP levels. Significant increases were also observed in mitochondrial LP and PC levels in AC group. Co-treatment with AC + CLA improved oxidative stress and mitochondrial dysfunction caused by AC. As a result of our findings, it was observed that CLA was effective in improving oxidative stress and impaired mitochondrial functions in brain tissue by the effect of AC. Considering the association between neurodegenerative diseases and mitochondrial dysfunction, CLA can play a role in the prevention and therapy of neurodegenerative disorders.
Collapse
Affiliation(s)
- Birsen Aydın
- Department of Biology, Faculty of Science, Amasya University, Amasya, Turkey
| | - Cansu Güler Şahin
- Department of Biology, Faculty of Science, Amasya University, Amasya, Turkey
| | - Vedat Şekeroğlu
- Department of Molecular Biology and Genetics, Faculty of Science, Ordu University, Ordu, Turkey
| | - Zülal Atlı Şekeroğlu
- Department of Molecular Biology and Genetics, Faculty of Science, Ordu University, Ordu, Turkey
| |
Collapse
|
12
|
Cristofano M D, A F, Giacomo M D, C F, F B, D L, Rotondi Aufiero V, F M, E C, G M, V Z, M R, P B. Mechanisms underlying the hormetic effect of conjugated linoleic acid: Focus on Nrf2, mitochondria and NADPH oxidases. Free Radic Biol Med 2021; 167:276-286. [PMID: 33753237 DOI: 10.1016/j.freeradbiomed.2021.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/17/2022]
Abstract
Nuclear factor erythroid 2-related factor2 (Nrf2) is a redox-sensitive transcription factor. Its activation by low dietary intake of ligands leads to antioxidant effects (eustress), while pro-oxidant effects (oxidative distress) may be associated with high doses. NADPH oxidases (NOXs) and the mitochondrial electron transport chain are the main sources of intracellular ROS, but their involvement in the biphasic/hormetic activity elicited by Nrf2 ligands is not fully understood. In this study, we investigated the involvement of NOX expression and mitochondrial function in the hormetic properties of omega-3 typically present in fish oil (FO) and conjugated linoleic acid (CLA) in the mouse liver. Four-week administration of FO, at both low and high doses (L-FO and H-FO) improves Nrf2-activated cyto-protection (by phase 2 enzymes), while a significant increase in respiration efficiency occurs in the liver mitochondria of H-FO BALB/c mice. Eustress conditions elicited by low dose CLA (L-CLA) are associated with increased activity of phase 2 enzymes, and with higher NOX1-2, mitochondrial defences, mitochondrial uncoupling protein 2 (UCP2), and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) expression, compared with controls. Steatogenic effects (lipid accumulation and alteration of lipid metabolism) elicited by high CLA (H-CLA) elicited that are associated with oxidative distress, increased mitochondrial complex I/III activity and reduced levels of phase 2 enzymes, in comparison with L-CLA-treated mice. Our results confirm the steatogenic activity of H-CLA and first demonstrate the role of NOX1 and NOX2 in the eustress conditions elicited by L-CLA. Notably, the negative association of the Nrf2/PGC-1α axis with the different CLA doses provides new insight into the mechanisms underlying the hormetic effect triggered by this Nrf2 ligand.
Collapse
Affiliation(s)
- Di Cristofano M
- Institute of Food Sciences, National Research Council (CNR-ISA), 83100, Avellino, Italy
| | - Ferramosca A
- Department of Environmental and Biological Sciences and Technologies, University of Salento, 73100, Lecce, Italy
| | - Di Giacomo M
- Department of Environmental and Biological Sciences and Technologies, University of Salento, 73100, Lecce, Italy
| | - Fusco C
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), 80100, Naples, Italy
| | - Boscaino F
- Institute of Food Sciences, National Research Council (CNR-ISA), 83100, Avellino, Italy
| | - Luongo D
- Institute of Food Sciences, National Research Council (CNR-ISA), 83100, Avellino, Italy
| | - Vera Rotondi Aufiero
- Institute of Food Sciences, National Research Council (CNR-ISA), 83100, Avellino, Italy
| | - Maurano F
- Institute of Food Sciences, National Research Council (CNR-ISA), 83100, Avellino, Italy
| | - Cocca E
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), 80100, Naples, Italy
| | - Mazzarella G
- Institute of Food Sciences, National Research Council (CNR-ISA), 83100, Avellino, Italy
| | - Zara V
- Department of Environmental and Biological Sciences and Technologies, University of Salento, 73100, Lecce, Italy
| | - Rossi M
- Institute of Food Sciences, National Research Council (CNR-ISA), 83100, Avellino, Italy
| | - Bergamo P
- Institute of Food Sciences, National Research Council (CNR-ISA), 83100, Avellino, Italy.
| |
Collapse
|
13
|
Mollica MP, Trinchese G, Cimmino F, Penna E, Cavaliere G, Tudisco R, Musco N, Manca C, Catapano A, Monda M, Bergamo P, Banni S, Infascelli F, Lombardi P, Crispino M. Milk Fatty Acid Profiles in Different Animal Species: Focus on the Potential Effect of Selected PUFAs on Metabolism and Brain Functions. Nutrients 2021; 13:1111. [PMID: 33800688 PMCID: PMC8066999 DOI: 10.3390/nu13041111] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/16/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
Milk contains several important nutrients that are beneficial for human health. This review considers the nutritional qualities of essential fatty acids (FAs), especially omega-3 (ω-3) and omega-6 (ω-6) polyunsaturated fatty acids (PUFAs) present in milk from ruminant and non-ruminant species. In particular, the impact of milk fatty acids on metabolism is discussed, including its effects on the central nervous system. In addition, we presented data indicating how animal feeding-the main way to modify milk fat composition-may have a potential impact on human health, and how rearing and feeding systems strongly affect milk quality within the same animal species. Finally, we have presented the results of in vivo studies aimed at supporting the beneficial effects of milk FA intake in animal models, and the factors limiting their transferability to humans were discussed.
Collapse
Affiliation(s)
- Maria P. Mollica
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.P.M.); (G.T.); (F.C.); (E.P.); (G.C.); (A.C.); (M.C.)
- BAT Center—Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples ‘Federico II’, 80055 Naples, Italy
| | - Giovanna Trinchese
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.P.M.); (G.T.); (F.C.); (E.P.); (G.C.); (A.C.); (M.C.)
- BAT Center—Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples ‘Federico II’, 80055 Naples, Italy
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.P.M.); (G.T.); (F.C.); (E.P.); (G.C.); (A.C.); (M.C.)
| | - Eduardo Penna
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.P.M.); (G.T.); (F.C.); (E.P.); (G.C.); (A.C.); (M.C.)
| | - Gina Cavaliere
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.P.M.); (G.T.); (F.C.); (E.P.); (G.C.); (A.C.); (M.C.)
| | - Raffaella Tudisco
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, 80100 Naples, Italy; (R.T.); (N.M.); (F.I.); (P.L.)
| | - Nadia Musco
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, 80100 Naples, Italy; (R.T.); (N.M.); (F.I.); (P.L.)
| | - Claudia Manca
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042 Cagliari, Italy; (C.M.); (S.B.)
| | - Angela Catapano
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.P.M.); (G.T.); (F.C.); (E.P.); (G.C.); (A.C.); (M.C.)
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Paolo Bergamo
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy
| | - Sebastiano Banni
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042 Cagliari, Italy; (C.M.); (S.B.)
| | - Federico Infascelli
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, 80100 Naples, Italy; (R.T.); (N.M.); (F.I.); (P.L.)
| | - Pietro Lombardi
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, 80100 Naples, Italy; (R.T.); (N.M.); (F.I.); (P.L.)
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.P.M.); (G.T.); (F.C.); (E.P.); (G.C.); (A.C.); (M.C.)
| |
Collapse
|
14
|
Cimmino F, Catapano A, Trinchese G, Cavaliere G, Culurciello R, Fogliano C, Penna E, Lucci V, Crispino M, Avallone B, Pizzo E, Mollica MP. Dietary Micronutrient Management to Treat Mitochondrial Dysfunction in Diet-Induced Obese Mice. Int J Mol Sci 2021; 22:2862. [PMID: 33799812 PMCID: PMC8000238 DOI: 10.3390/ijms22062862] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/25/2021] [Accepted: 03/08/2021] [Indexed: 02/04/2023] Open
Abstract
Obesity and associated metabolic disturbances, which have been increasing worldwide in recent years, are the consequences of unhealthy diets and physical inactivity and are the main factors underlying non-communicable diseases (NCD). These diseases are now responsible for about three out of five deaths worldwide, and it has been shown that they depend on mitochondrial dysfunction, systemic inflammation and oxidative stress. It was also demonstrated that several nutritional components modulating these processes are able to influence metabolic homeostasis and, consequently, to prevent or delay the onset of NCD. An interesting combination of nutraceutical substances, named DMG-gold, has been shown to promote metabolic and physical wellness. The aim of this research was to investigate the metabolic, inflammatory and oxidative pathways modulated by DMG-gold in an animal model with diet-induced obesity. Our data indicate that DMG-gold decreases the metabolic efficiency and inflammatory state and acts as an antioxidant and detoxifying agent, modulating mitochondrial functions. Therefore, DMG-gold is a promising candidate in the prevention/treatment of NCD.
Collapse
Affiliation(s)
- Fabiano Cimmino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
| | - Angela Catapano
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Giovanna Trinchese
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
| | - Gina Cavaliere
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
| | - Rosanna Culurciello
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
| | - Chiara Fogliano
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
| | - Eduardo Penna
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
| | - Valeria Lucci
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
- IEOS, Institute of Experimental Endocrinology and Oncology “G. Salvatore”—National Research Council, 80131 Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
| | - Bice Avallone
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
| | - Elio Pizzo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (F.C.); (A.C.); (G.T.); (G.C.); (R.C.); (C.F.); (E.P.); (V.L.); (M.C.); (B.A.); (E.P.)
| |
Collapse
|
15
|
Modulatory role of dietary polyunsaturated fatty acids in Nrf2-mediated redox homeostasis. Prog Lipid Res 2020; 80:101066. [DOI: 10.1016/j.plipres.2020.101066] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
|
16
|
Islam MA, Khandker SS, Kotyla PJ, Hassan R. Immunomodulatory Effects of Diet and Nutrients in Systemic Lupus Erythematosus (SLE): A Systematic Review. Front Immunol 2020; 11:1477. [PMID: 32793202 PMCID: PMC7387408 DOI: 10.3389/fimmu.2020.01477] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multiple organ involvement, including the skin, joints, kidneys, lungs, central nervous system and the haematopoietic system, with a large number of complications. Despite years of study, the etiology of SLE remains unclear; thus, safe and specifically targeted therapies are lacking. In the last 20 years, researchers have explored the potential of nutritional factors on SLE and have suggested complementary treatment options through diet. This study systematically reviews and evaluates the clinical and preclinical scientific evidence of diet and dietary supplementation that either alleviate or exacerbate the symptoms of SLE. For this review, a systematic literature search was conducted using PubMed, Scopus and Google Scholar databases only for articles written in the English language. Based on the currently published literature, it was observed that a low-calorie and low-protein diet with high contents of fiber, polyunsaturated fatty acids, vitamins, minerals and polyphenols contain sufficient potential macronutrients and micronutrients to regulate the activity of the overall disease by modulating the inflammation and immune functions of SLE.
Collapse
Affiliation(s)
- Md Asiful Islam
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Shahad Saif Khandker
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Dhaka, Bangladesh
| | - Przemysław J Kotyla
- Department of Internal Medicine, Rheumatology and Clinical Immunology, Medical Faculty in Katowice, Medical University of Silesia, Katowice, Poland
| | - Rosline Hassan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
17
|
Luongo D, Treppiccione L, Maurano F, Rossi M, Bergamo P. The murine enterocyte cell line Mode-K is a novel and reliable in vitro model for studies on gluten toxicity. Food Chem Toxicol 2020; 140:111331. [DOI: 10.1016/j.fct.2020.111331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/26/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022]
|
18
|
Annunziata C, Lama A, Pirozzi C, Cavaliere G, Trinchese G, Di Guida F, Nitrato Izzo A, Cimmino F, Paciello O, De Biase D, Murru E, Banni S, Calignano A, Mollica MP, Mattace Raso G, Meli R. Palmitoylethanolamide counteracts hepatic metabolic inflexibility modulating mitochondrial function and efficiency in diet-induced obese mice. FASEB J 2019; 34:350-364. [PMID: 31914699 DOI: 10.1096/fj.201901510rr] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR)-α activation controls hepatic lipid homeostasis, stimulating fatty acid oxidation, and adapting the metabolic response to lipid overload and storage. Here, we investigate the effect of palmitoylethanolamide (PEA), an endogenous PPAR-α ligand, in counteracting hepatic metabolic inflexibility and mitochondrial dysfunction induced by high-fat diet (HFD) in mice. Long-term PEA administration (30 mg/kg/die per os) in HFD mice limited hepatic lipid accumulation, increased energy expenditure, and markedly reduced insulin resistance. In isolated liver mitochondria, we have demonstrated PEA capability to modulate mitochondrial oxidative capacity and energy efficiency, leading to the reduction of intracellular lipid accumulation and oxidative stress. Moreover, we have evaluated the effect of PEA on mitochondrial bioenergetics of palmitate-challenged HepG2 cells, using Seahorse analyzer. In vitro data showed that PEA recovered mitochondrial dysfunction and reduced lipid accumulation in insulin-resistant HepG2 cells, increasing fatty acid oxidation. Mechanistic studies showed that PEA effect on lipid metabolism was limited by AMP-activated protein kinase (AMPK) inhibition, providing evidence for a pivotal role of AMPK in PEA-induced adaptive metabolic setting. All these findings identify PEA as a modulator of hepatic lipid and glucose homeostasis, limiting metabolic inflexibility induced by nutrient overload.
Collapse
Affiliation(s)
- Chiara Annunziata
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Adriano Lama
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gina Cavaliere
- Department of Biology, University of Naples Federico II, Naples, Italy
| | | | | | | | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Davide De Biase
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Elisabetta Murru
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Cagliari, Italy
| | - Sebastiano Banni
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Cagliari, Italy
| | - Antonio Calignano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | | | - Rosaria Meli
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
19
|
Cigliano L, Spagnuolo MS, Boscaino F, Ferrandino I, Monaco A, Capriello T, Cocca E, Iannotta L, Treppiccione L, Luongo D, Maurano F, Rossi M, Bergamo P. Dietary Supplementation with Fish Oil or Conjugated Linoleic Acid Relieves Depression Markers in Mice by Modulation of the Nrf2 Pathway. Mol Nutr Food Res 2019; 63:e1900243. [DOI: 10.1002/mnfr.201900243] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/25/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Luisa Cigliano
- Department of BiologyUniversity “Federico II” of Naples Naples Italy
| | - Maria Stefania Spagnuolo
- Institute for Animal Production System in Mediterranean EnvironmentNational Research Council (ISPAAM, CNR) Naples Italy
| | - Floriana Boscaino
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| | - Ida Ferrandino
- Department of BiologyUniversity “Federico II” of Naples Naples Italy
| | - Antonio Monaco
- Department of BiologyUniversity “Federico II” of Naples Naples Italy
| | - Teresa Capriello
- Department of BiologyUniversity “Federico II” of Naples Naples Italy
| | - Ennio Cocca
- Institute of Biosciences and Bio‐ResourcesNational Research Council (CNR‐IBBR) Naples Italy
| | - Lucia Iannotta
- Department of BiologyUniversity “Federico II” of Naples Naples Italy
| | - Lucia Treppiccione
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| | - Diomira Luongo
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| | - Francesco Maurano
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| | - Mauro Rossi
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| | - Paolo Bergamo
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| |
Collapse
|
20
|
Trinchese G, Cavaliere G, Penna E, De Filippo C, Cimmino F, Catapano A, Musco N, Tudisco R, Lombardi P, Infascelli F, Messina G, Muredda L, Banni S, Monda M, Crispino M, Mollica MP. Milk From Cow Fed With High Forage/Concentrate Ratio Diet: Beneficial Effect on Rat Skeletal Muscle Inflammatory State and Oxidative Stress Through Modulation of Mitochondrial Functions and AMPK Activity. Front Physiol 2019; 9:1969. [PMID: 30705640 PMCID: PMC6344429 DOI: 10.3389/fphys.2018.01969] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 12/31/2018] [Indexed: 12/25/2022] Open
Abstract
Milk and dairy products are relevant components of daily diet and are part of dietary recommendation in many countries due to their content of key nutrients. However, the relatively high content of saturated fat of the milk and its extensive usage for every age group raises concerns about its potential negative health effects. Therefore, in the last years, several researchers dedicated their attention to milk production and quality. Milk fatty acids profile depend on cow feeding and in particular on the type of forage and concentrate and forage/concentrate ratio. It was demonstrated that feeding dairy cows with a 70/30 forage/concentrate ratio yields milk with a low ω6:ω3 ratio and high CLA levels. In this work, we demonstrated that the supplementation of rats diet with this high forage milk (HFM) results, in the skeletal muscle of these animals, in a reduced lipid content and inflammation levels, and an improved mitochondrial lipid oxidation, and redox status through modulation of AMPK activity.
Collapse
Affiliation(s)
| | - Gina Cavaliere
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Eduardo Penna
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Chiara De Filippo
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Angela Catapano
- Department of Biology, University of Naples Federico II, Naples, Italy.,Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Nadia Musco
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Raffaella Tudisco
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Pietro Lombardi
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Federico Infascelli
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Laura Muredda
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Sebastiano Banni
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
21
|
Queiroz MP, Lima MDS, de Melo MFFT, Bertozzo CCDMS, de Araújo DF, Guerra GCB, Queiroga RDCRDE, Soares JKB. Maternal suppplementation with conjugated linoleic acid reduce anxiety and lipid peroxidation in the offspring brain. J Affect Disord 2019; 243:75-82. [PMID: 30236761 DOI: 10.1016/j.jad.2018.09.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 08/09/2018] [Accepted: 09/10/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Maternal consumption of fatty acids can alter neuronal membrane function, synaptic connections, and protect the brain from alterations caused by disturbances such as lipid peroxidation and anxiety in the offspring. We aimed to investigate how the maternal consumption of conjugated linoleic acid (CLA) interferes in anxiety behavior of the offspring and cerebral lipid peroxidation. METHODS Three groups were formed: control (CG) - diet without CLA; CLA1 - diet containing 1% of CLA; and CLA3 - diet containing 3% of CLA. These diets were offered to the mothers from the 7th day of gestation until the end of lactation. The following behavioral tests were used: Elevated plus maze (EPM), Open Field (OF) and Light-dark Box (LDB). Levels of malondialdehyde (MDA) and glutathione were measured in the offspring's brains. Data were analyzed by ANOVA followed by the Holm-Sidak post-test or the Kruskal-Wallis test (p < 0.05). RESULTS CLA1 and CLA3 showed higher number of entries in the open arms and time spent in the central area in EPM, they translocated and ambulated more in the clear area of the LDB and presented more rearing in the OF compared to CG (p < 0.05); moreover, they presented higher concentration of glutathione and lower MDA in brain tissue (p < 0.05). LIMITATIONS We evaluated the effect of maternal consumption of CLA on anxiety and lipid peroxidation in rats' offspring, but a similar study should be performed in humans. CONCLUSIONS Maternal intake of CLA induced a decrease in the parameters of anxiety and cerebral lipid peroxidation in the offspring.
Collapse
Affiliation(s)
- Michelly Pires Queiroz
- Program of Food Science and Tecnology, Federal University of Paraiba, Cidade Universitária, s/n - Castelo Branco III, João Pessoa, PB 58051-085, Brazil.
| | - Martiniano da Silva Lima
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, CG, Brazil
| | | | | | | | | | - Rita de Cassia Ramos do Egypto Queiroga
- Program of Food Science and Tecnology, Federal University of Paraiba, Cidade Universitária, s/n - Castelo Branco III, João Pessoa, PB 58051-085, Brazil; Laboratory of Bromatology, Department of Nutrition, Federal University of Paraiba, João Pessoa, PB, Brazil.
| | - Juliana Késsia Barbosa Soares
- Program of Food Science and Tecnology, Federal University of Paraiba, Cidade Universitária, s/n - Castelo Branco III, João Pessoa, PB 58051-085, Brazil; Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, CG, Brazil
| |
Collapse
|
22
|
Qi XL, Wang J, Yue HY, Wu SG, Zhang YN, Ni HM, Guo Y, Zhang HJ, Qi GH. Trans10, cis12-conjugated linoleic acid exhibits a stronger antioxidant capacity than cis9, trans11-conjugated linoleic acid in primary cultures of laying hen hepatocytes. Poult Sci 2018; 97:4415-4424. [DOI: 10.3382/ps/pey297] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 06/20/2018] [Indexed: 12/23/2022] Open
|
23
|
Cavaliere G, Viggiano E, Trinchese G, De Filippo C, Messina A, Monda V, Valenzano A, Cincione RI, Zammit C, Cimmino F, Catapano A, Sessa F, Messina G, Monda M, Crispino M, Mollica MP. Long Feeding High-Fat Diet Induces Hypothalamic Oxidative Stress and Inflammation, and Prolonged Hypothalamic AMPK Activation in Rat Animal Model. Front Physiol 2018; 9:818. [PMID: 30034345 PMCID: PMC6043859 DOI: 10.3389/fphys.2018.00818] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/11/2018] [Indexed: 12/14/2022] Open
Abstract
Scope: The hypothalamus is a key brain region involved in the control of feeding and energy expenditure. Hypothalamic inflammation and oxidative stress are landmarks of both obesity and aging processes, although the molecular mechanisms are still unknown. Therefore, with the aim to understand the neurobiological mechanisms of energy homeostasis during aging, we evaluate the effects of long feeding high-fat diet (HFD) in rats, at different age, on modulation of hypothalamic molecular pathway, oxidative stress, and inflammation. Procedures: Male Wistar rats were divided into two groups: control group, receiving standard diet (CD), and treated group, receiving HFD. Both groups were treated with the appropriate diet for 1, 3, 6, 12, or 18 weeks. We investigated energy balance and body composition, as well as lipid profile, homeostatic model assessment index, and inflammatory state in serum. Furthermore, we also analyzed, at hypothalamic level, inflammation and oxidative stress, and adenosine monophosphate-dependent kinase (AMPK) and pAMPK expression levels. Results: Our data showed that aging and HFD induce increased energy intake and energy efficiency and decreased energy expenditure associated, at hypothalamic level, with inflammation and oxidative stress and activation of AMPK. Conclusion: Our results indicate that the age at which HFD feeding starts and the diet duration are critical in obesity development. The prolonged activation of hypothalamic AMPK may be related to the alterations in energy homeostasis.
Collapse
Affiliation(s)
- Gina Cavaliere
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Emanuela Viggiano
- Section of Human Physiology, Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy.,Prenatal Medicine, ULSS6 Euganea, Padua, Italy
| | | | - Chiara De Filippo
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Antonietta Messina
- Unit of Dietetics and Sports Medicine, Section of Human Physiology, Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Vincenzo Monda
- Unit of Dietetics and Sports Medicine, Section of Human Physiology, Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Raffaele I Cincione
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | | | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Angela Catapano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Francesco Sessa
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Marcellino Monda
- Unit of Dietetics and Sports Medicine, Section of Human Physiology, Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
24
|
Trinchese G, Cavaliere G, De Filippo C, Aceto S, Prisco M, Chun JT, Penna E, Negri R, Muredda L, Demurtas A, Banni S, Berni-Canani R, Mattace Raso G, Calignano A, Meli R, Greco L, Crispino M, Mollica MP. Human Milk and Donkey Milk, Compared to Cow Milk, Reduce Inflammatory Mediators and Modulate Glucose and Lipid Metabolism, Acting on Mitochondrial Function and Oleylethanolamide Levels in Rat Skeletal Muscle. Front Physiol 2018; 9:32. [PMID: 29472867 PMCID: PMC5810302 DOI: 10.3389/fphys.2018.00032] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 01/10/2018] [Indexed: 12/15/2022] Open
Abstract
Scope: Milk from various species differs in nutrient composition. In particular, human milk (HM) and donkey milk (DM) are characterized by a relative high level of triacylglycerol enriched in palmitic acid in sn-2 position. These dietary fats seem to exert beneficial nutritional properties through N-acylethanolamine tissue modulation. The aim of this study is to compare the effects of cow milk (CM), DM, and HM on inflammation and glucose and lipid metabolism, focusing on mitochondrial function, efficiency, and dynamics in skeletal muscle, which is the major determinant of resting metabolic rate. Moreover, we also evaluated the levels of endocannabinoids and N-acylethanolamines in liver and skeletal muscle, since tissue fatty acid profiles can be modulated by nutrient intervention. Procedures: To this aim, rats were fed with CM, DM, or HM for 4 weeks. Then, glucose tolerance and insulin resistance were analyzed. Pro-inflammatory and anti-inflammatory cytokines were evaluated in serum and skeletal muscle. Skeletal muscle was also processed to estimate mitochondrial function, efficiency, and dynamics, oxidative stress, and antioxidant/detoxifying enzyme activities. Fatty acid profiles, endocannabinoids, and N-acylethanolamine congeners were determined in liver and skeletal muscle tissue. Results: We demonstrated that DM or HM administration reducing inflammation status, improves glucose disposal and insulin resistance and reduces lipid accumulation in skeletal muscle. Moreover, HM or DM administration increases redox status, and mitochondrial uncoupling, affecting mitochondrial dynamics in the skeletal muscle. Interestingly, HM and DM supplementation increase liver and muscle levels of the N-oleoylethanolamine (OEA), a key regulator of lipid metabolism and inflammation. Conclusions: HM and DM have a healthy nutritional effect, acting on inflammatory factors and glucose and lipid metabolism. This beneficial effect is associated to a modulation of mitochondrial function, efficiency, and dynamics and to an increase of OEA levels in skeletal muscle.
Collapse
Affiliation(s)
| | - Gina Cavaliere
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Chiara De Filippo
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Serena Aceto
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Marina Prisco
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Jong Tai Chun
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Eduardo Penna
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Rossella Negri
- European Laboratory for Food Induced Diseases, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Laura Muredda
- Dipartimento di Scienze Biomediche, Università degli Studi di Cagliari, Cagliari, Italy
| | - Andrea Demurtas
- Dipartimento di Scienze Biomediche, Università degli Studi di Cagliari, Cagliari, Italy
| | - Sebastiano Banni
- Dipartimento di Scienze Biomediche, Università degli Studi di Cagliari, Cagliari, Italy
| | - Roberto Berni-Canani
- European Laboratory for Food Induced Diseases, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | | | - Antonio Calignano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Rosaria Meli
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Luigi Greco
- European Laboratory for Food Induced Diseases, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Maria P Mollica
- Department of Biology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
25
|
Monaco A, Ferrandino I, Boscaino F, Cocca E, Cigliano L, Maurano F, Luongo D, Spagnuolo MS, Rossi M, Bergamo P. Conjugated linoleic acid prevents age-dependent neurodegeneration in a mouse model of neuropsychiatric lupus via the activation of an adaptive response. J Lipid Res 2018; 59:48-57. [PMID: 29167408 PMCID: PMC5748496 DOI: 10.1194/jlr.m079400] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/14/2017] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress is a key mediator of autoimmune/neurodegenerative disorders. The antioxidant/anti-inflammatory effect of a synthetic conjugated linoleic acid (CLA) mixture in MRL/MpJ-Fas lpr mice (MRL/lpr), an animal model of neuropsychiatric lupus, was previously associated with the improvement of nuclear factor-E2-related factor 2 (Nrf2) defenses in the spleen and liver. However, little is known about the neuroprotective ability of a CLA mixture. This study investigated the age-dependent progression of oxidative stress and the hyperactivation of redox-sensitive compensatory pathways (macroautophagy, Nrf2) in old/diseased MRL/lpr mice brains and examines the effect produced by dietary CLA supplementation. Disrupted redox homeostasis was evidenced in the blood, liver, and brain of 21- to 22-week-old MRL/lpr (Old) mice compared with 8- to 10-week-old MRL/lpr (Young) animals. This alteration was associated with significant hyperactivation of compensatory mechanisms (macroautophagy, Nrf2, and astrocyte activation) in the brains of Old mice. Five-week daily supplementation with CLA (650 mg/kg-1 body weight) of 16-week-old (CLA+Old) mice diminished all the pathological hallmarks at a level comparable to Young mice or healthy controls (BALB/c). Such data demonstrated that MRL/lpr mice can serve as a valuable model for the evaluation of the effectiveness of neuroprotective drugs. Notably, the preventive effect provided by CLA supplementation against age-associated neuronal damage and hyperactivation of compensatory mechanisms suggests that the activation of an adaptive response is at least in part accountable for its neuroprotective ability.
Collapse
Affiliation(s)
- Antonio Monaco
- Department of Biology, University "Federico II" of Naples, Italy
| | - Ida Ferrandino
- Department of Biology, University "Federico II" of Naples, Italy
| | - Floriana Boscaino
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Ennio Cocca
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), Naples, Italy
| | - Luisa Cigliano
- Department of Biology, University "Federico II" of Naples, Italy
| | - Francesco Maurano
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Diomira Luongo
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Maria Stefania Spagnuolo
- Institute for Animal Production System in Mediterranean Environment, National Research Council (ISPAAM, CNR) Naples, Italy
| | - Mauro Rossi
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Paolo Bergamo
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| |
Collapse
|
26
|
Barbosa MQ, Queiroga RDCRE, Bertozzo CCDMS, Araújo DFDS, Oliveira LIG, Silva JYP, Bomfim MAD, Guerra GCB, Costa S, Bessa R, Alves S, Barbosa Soares JK. Effect of diets with goat milk fat supplemented with exercise on anxiety and oxidative stress in the brains of adult rats. Food Funct 2018; 9:2891-2901. [DOI: 10.1039/c7fo01764b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Goat milk fat induced anxiolytic effect in sedentary animals; exercise promoted lipid peroxidation in the brain; exercise induced anxiety.
Collapse
|
27
|
Winberg LK, Jacobsen S, Nielsen CH. Microparticles from patients with systemic lupus erythematosus induce production of reactive oxygen species and degranulation of polymorphonuclear leukocytes. Arthritis Res Ther 2017; 19:230. [PMID: 29041967 PMCID: PMC5646131 DOI: 10.1186/s13075-017-1437-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/20/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The interaction of circulating microparticles (MPs) with immune cells in systemic lupus erythematosus (SLE) is sparsely investigated. We examined the ability of MPs from SLE patients to induce production of reactive oxygen species (ROS) and degranulation of polymorphonuclear leukocytes (PMNs). METHODS Plasma MPs, leukocytes and sera isolated from 20 SLE patients and 10 healthy controls were mixed in different combinations, with or without lipopolysaccharide (LPS), and incubated for 30 min. Dihydrorhodamine 123 was used to measure ROS production by flow cytometry. The ability of immunoglobulin G (IgG) isolated from five SLE patients to increase MP-induced production of ROS by PMNs was tested. Cell supernatants were analysed for content of primary, secondary and tertiary granule components by Luminex assays. RESULTS MPs from SLE patients promoted ROS production by PMNs, and enhanced LPS-induced ROS production and release of primary granules by PMNs, when added to samples of autologous leukocytes and serum. In a similar autologous setting, MPs from healthy controls enhanced LPS-induced ROS production by PMNs. When leukocytes from a healthy control were stimulated with autologous MPs in the presence of various sera, SLE patient serum promoted ROS production and release of primary and secondary granules by PMNs. A role for antibodies in this respect was indicated by the observation that supplementation of normal serum with IgG from SLE patients promoted MP-induced ROS production by healthy PMNs. Moreover, when various MPs were incubated with leukocytes and serum from a healthy control, patient-derived MPs induced more ROS production by PMNs than did healthy control-derived MPs. CONCLUSIONS SLE patients display increased ROS production and degranulation by PMNs in response to MPs, which partly depends on serum components, including antibodies, MP properties and hyper-responsiveness of the PMNs per se.
Collapse
Affiliation(s)
- Line Kjær Winberg
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Section 7521, Copenhagen, Denmark.,Copenhagen Lupus and Vasculitis Clinic, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Section 4242, Blegdamsvej 9, DK-2100, Copenhagen, Denmark
| | - Søren Jacobsen
- Copenhagen Lupus and Vasculitis Clinic, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Section 4242, Blegdamsvej 9, DK-2100, Copenhagen, Denmark
| | - Claus H Nielsen
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Section 7521, Copenhagen, Denmark.
| |
Collapse
|
28
|
Aydın B, Atlı Şekeroğlu Z, Şekeroğlu V. Acrolein-induced oxidative stress and genotoxicity in rats: protective effects of whey protein and conjugated linoleic acid. Drug Chem Toxicol 2017; 41:225-231. [DOI: 10.1080/01480545.2017.1354872] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Birsen Aydın
- Department of Biology, Faculty of Science, Amasya University, Amasya, Turkey
| | - Zülal Atlı Şekeroğlu
- Department of Molecular Biology and Genetics, Faculty of Science, Ordu University, Ordu, Turkey
| | - Vedat Şekeroğlu
- Department of Molecular Biology and Genetics, Faculty of Science, Ordu University, Ordu, Turkey
| |
Collapse
|
29
|
Mollica MP, Mattace Raso G, Cavaliere G, Trinchese G, De Filippo C, Aceto S, Prisco M, Pirozzi C, Di Guida F, Lama A, Crispino M, Tronino D, Di Vaio P, Berni Canani R, Calignano A, Meli R. Butyrate Regulates Liver Mitochondrial Function, Efficiency, and Dynamics in Insulin-Resistant Obese Mice. Diabetes 2017; 66:1405-1418. [PMID: 28223285 DOI: 10.2337/db16-0924] [Citation(s) in RCA: 238] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 02/14/2017] [Indexed: 12/15/2022]
Abstract
Fatty liver, oxidative stress, and mitochondrial dysfunction are key pathophysiological features of insulin resistance and obesity. Butyrate, produced by fermentation in the large intestine by gut microbiota, and its synthetic derivative, the N-(1-carbamoyl-2-phenyl-ethyl) butyramide, FBA, have been demonstrated to be protective against insulin resistance and fatty liver. Here, hepatic mitochondria were identified as the main target of the beneficial effect of both butyrate-based compounds in reverting insulin resistance and fat accumulation in diet-induced obese mice. In particular, butyrate and FBA improved respiratory capacity and fatty acid oxidation, activated the AMPK-acetyl-CoA carboxylase pathway, and promoted inefficient metabolism, as shown by the increase in proton leak. Both treatments consistently increased utilization of substrates, especially fatty acids, leading to the reduction of intracellular lipid accumulation and oxidative stress. Finally, the shift of the mitochondrial dynamic toward fusion by butyrate and FBA resulted in the improvement not only of mitochondrial cell energy metabolism but also of glucose homeostasis. In conclusion, butyrate and its more palatable synthetic derivative, FBA, modulating mitochondrial function, efficiency, and dynamics, can be considered a new therapeutic strategy to counteract obesity and insulin resistance.
Collapse
Affiliation(s)
| | | | - Gina Cavaliere
- Department of Biology, University of Naples Federico II, Naples, Italy
| | | | - Chiara De Filippo
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Serena Aceto
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Marina Prisco
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | - Adriano Lama
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Diana Tronino
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Paola Di Vaio
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- European Laboratory for Investigation of Food Induced Diseases, University of Naples Federico II, Naples, Italy
- CEINGE Advanced Biotechnology, University of Naples Federico II, Naples, Italy
| | - Antonio Calignano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Rosaria Meli
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
30
|
Abstract
AbstractSystemic lupus erythematosus (SLE) is a chronic inflammatory and autoimmune disease characterised by multiple organ involvement and a large number of complications. SLE management remains complicated owing to the biological heterogeneity between patients and the lack of safe and specific targeted therapies. There is evidence that dietary factors can contribute to the geoepidemiology of autoimmune diseases such as SLE. Thus, diet therapy could be a promising approach in SLE owing to both its potential prophylactic effects, without the side effects of classical pharmacology, and its contribution to reducing co-morbidities and improving quality of life in patients with SLE. However, the question arises as to whether nutrients could ameliorate or exacerbate SLE and how they could modulate inflammation and immune function at a molecular level. The present review summarises preclinical and clinical experiences to provide the reader with an update of the positive and negative aspects of macro- and micronutrients and other nutritional factors, including dietary phenols, on SLE, focusing on the mechanisms of action involved.
Collapse
|
31
|
Konopka AR, Laurin JL, Musci RV, Wolff CA, Reid JJ, Biela LM, Zhang Q, Peelor FF, Melby CL, Hamilton KL, Miller BF. Influence of Nrf2 activators on subcellular skeletal muscle protein and DNA synthesis rates after 6 weeks of milk protein feeding in older adults. GeroScience 2017; 39:175-186. [PMID: 28283797 DOI: 10.1007/s11357-017-9968-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/02/2017] [Indexed: 12/12/2022] Open
Abstract
In older adults, chronic oxidative and inflammatory stresses are associated with an impaired increase in skeletal muscle protein synthesis after acute anabolic stimuli. Conjugated linoleic acid (CLA) and Protandim have been shown to activate nuclear factor erythroid-derived 2-like 2 (Nrf2), a transcription factor for the antioxidant response element and anti-inflammatory pathways. This study tested the hypothesis that compared to a placebo control (CON), CLA and Protandim would increase skeletal muscle subcellular protein (myofibrillar, mitochondrial, cytoplasmic) and DNA synthesis in older adults after 6 weeks of milk protein feeding. CLA decreased oxidative stress and skeletal muscle oxidative damage with a trend to increase messenger RNA (mRNA) expression of a Nrf2 target, NAD(P)H dehydrogenase quinone 1 (NQO1). However, CLA did not influence other Nrf2 targets (heme oxygenase-1 (HO-1), glutathione peroxidase 1 (Gpx1)) or protein or DNA synthesis. Conversely, Protandim increased HO-1 protein content but not the mRNA expression of downstream Nrf2 targets, oxidative stress, or skeletal muscle oxidative damage. Rates of myofibrillar protein synthesis were maintained despite lower mitochondrial and cytoplasmic protein syntheses after Protandim versus CON. Similarly, DNA synthesis was non-significantly lower after Protandim compared to CON. After Protandim, the ratio of protein to DNA synthesis tended to be greater in the myofibrillar fraction and maintained in the mitochondrial and cytoplasmic fractions, emphasizing the importance of measuring both protein and DNA synthesis to gain insight into proteostasis. Overall, these data suggest that Protandim may enhance proteostatic mechanisms of skeletal muscle contractile proteins after 6 weeks of milk protein feeding in older adults.
Collapse
Affiliation(s)
- Adam R Konopka
- Department of Health and Exercise Science, Colorado State University, 110 Moby B Complex, Fort Collins, CO, 80523, USA.
| | - Jaime L Laurin
- Department of Health and Exercise Science, Colorado State University, 110 Moby B Complex, Fort Collins, CO, 80523, USA
| | - Robert V Musci
- Department of Health and Exercise Science, Colorado State University, 110 Moby B Complex, Fort Collins, CO, 80523, USA
| | - Christopher A Wolff
- Department of Health and Exercise Science, Colorado State University, 110 Moby B Complex, Fort Collins, CO, 80523, USA
| | - Justin J Reid
- Department of Health and Exercise Science, Colorado State University, 110 Moby B Complex, Fort Collins, CO, 80523, USA
| | - Laurie M Biela
- Department of Health and Exercise Science, Colorado State University, 110 Moby B Complex, Fort Collins, CO, 80523, USA
| | - Qian Zhang
- Department of Health and Exercise Science, Colorado State University, 110 Moby B Complex, Fort Collins, CO, 80523, USA
| | - Fredrick F Peelor
- Department of Health and Exercise Science, Colorado State University, 110 Moby B Complex, Fort Collins, CO, 80523, USA
| | - Christopher L Melby
- Department of Health and Exercise Science, Colorado State University, 110 Moby B Complex, Fort Collins, CO, 80523, USA
| | - Karyn L Hamilton
- Department of Health and Exercise Science, Colorado State University, 110 Moby B Complex, Fort Collins, CO, 80523, USA
| | - Benjamin F Miller
- Department of Health and Exercise Science, Colorado State University, 110 Moby B Complex, Fort Collins, CO, 80523, USA
| |
Collapse
|
32
|
Luongo D, Coppola A, Treppiccione L, Bergamo P, Sorrentino A, Ferrocino I, Turroni S, Neviani E, Di Cagno R, Cocolin L, Rossi M. Modulation of the cytokine profile in Caco-2 cells by faecal lactobacilli and bifidobacteria from individuals with distinct dietary habits. Cytokine 2017; 90:80-87. [PMID: 27863334 DOI: 10.1016/j.cyto.2016.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 10/16/2016] [Accepted: 11/10/2016] [Indexed: 12/18/2022]
Abstract
Enterocytes are actively involved in the defense against pathogens and they limit penetration of commensal microbes into tissues. They also have an important role in gut immunity as enterocytes confer mucosal dendritic cell specialisation. On the other hand, the microbiota is directly involved in the development and modulation of the intestinal immune system. Particularly, lactobacilli and bifidobacteria play a primary role in shaping the immune response. We further explored this issue by evaluating whether functional differences in Caco-2 cells could characterise faecal populations of lactobacilli (155 samples) and bifidobacteria (110 samples) isolated from three dietary cohorts (omnivores, ovo-lacto-vegetarians and vegans) recruited at four Italian centres (Turin, Parma, Bologna and Bari). According to our findings, tested bacteria were unable to modulate expression of IL-8, IL-10, TGF-β or thymic stromal lymphopoietin (TSLP) cytokines in unstimulated Caco-2 cells. Conversely, in phorbol 12-myristate 13-acetate and ionomycin (PMA/Io) stimulated Caco-2 cells, lactobacilli from the omnivorous group and all bifidobacteria significantly down-regulated IL-8. Notably, both genera also lowered the TSLP expression in stimulated Caco-2 cells, regardless of the diet regimen. By further examining these data on the basis of geographical origin, we found that lactobacilli from the vegetarian group recruited in Bari, significantly up-regulated this cytokine. In conclusion, we highlighted a peculiar immune-modulatory activity profile for lactobacilli on enterocytes undergoing a stimulatory signal, which was associated with a specific dietary habit. Furthermore, the geographical area had a significant impact on the inflammatory potential of members of the Lactobacillus genus.
Collapse
Affiliation(s)
| | | | | | | | | | - Ilario Ferrocino
- Department of Agricultural, Forest and Food Science, University of Turin, Grugliasco, Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Erasmo Neviani
- Laboratory of Food Microbiology, Department of Food Science, University of Parma, Parco Area delle Scienze 48/A, Parma, Italy
| | - Raffaella Di Cagno
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Luca Cocolin
- Department of Agricultural, Forest and Food Science, University of Turin, Grugliasco, Italy
| | - Mauro Rossi
- Institute of Food Sciences, CNR, Avellino, Italy.
| |
Collapse
|
33
|
Viggiano E, Mollica MP, Lionetti L, Cavaliere G, Trinchese G, De Filippo C, Chieffi S, Gaita M, Barletta A, De Luca B, Crispino M, Monda M. Effects of an High-Fat Diet Enriched in Lard or in Fish Oil on the Hypothalamic Amp-Activated Protein Kinase and Inflammatory Mediators. Front Cell Neurosci 2016; 10:150. [PMID: 27375435 PMCID: PMC4899473 DOI: 10.3389/fncel.2016.00150] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/27/2016] [Indexed: 01/10/2023] Open
Abstract
The high fat diet (HFD) rich in lard induces obesity, inflammation and oxidative stress, and the deregulation of hypothalamic nuclei plays an important role in this mechanism. One important factor involved in the food intake and inflammation is adenosine monophosphate-dependent kinase (AMPK), a serine/threonine kinase activated by phosphorylation. Omega (ω)3-polyunsaturated fatty acids (PUFA) are dietary compounds known to attenuate the obesity-related diseases, although the molecular mechanisms underlying their actions in the hypothalamus are not completely understood. We hypothesized that the beneficial effects of PUFA may be mediated by AMPK in the hypothalamus. To this aim, rats were fed a control diet (CD), or isocaloric HFD containing either fish oil (FD; rich in ω3-PUFA) or lard for 6 weeks, and the activation of AMPK, inflammatory state (IKKβ, TNF-α) and oxidative stress were analyzed in the hypothalamus. In addition, we also studied serum lipid profile, homeostatic model assessment (HOMA) index, and pro-inflammatory parameters. Our results showed, at the hypothalamic level of LD-fed rats, an increase of AMPK activation, inflammation and oxidative stress, while no modifications were detected in FD-fed animals compared to CD. In addition body weight gain, serum lipid profile, pro-inflammatory parameters and insulin resistance were reduced in FD animals compared to LD. In conclusion, our data indicate that the substitution of saturated by unsaturated fatty acids in the diet has beneficial effects on modulation of hypothalamic inflammation and function in obesity, underlying, at hypothalamic level, the interaction among insulin and/or leptin resistance, AMPK activation and hyperphagia.
Collapse
Affiliation(s)
- Emanuela Viggiano
- Department of Experimental Medicine-Section of Human Physiology, Second University of NaplesNaples, Italy; Department of Medicine, University of PadovaPadua, Italy
| | | | - Lillà Lionetti
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Gina Cavaliere
- Department of Biology, University of Naples Federico II Naples, Italy
| | | | - Chiara De Filippo
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Sergio Chieffi
- Department of Experimental Medicine-Section of Human Physiology, Second University of Naples Naples, Italy
| | - Marcello Gaita
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Antonio Barletta
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Bruno De Luca
- Department of Experimental Medicine-Section of Human Physiology, Second University of Naples Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine-Section of Human Physiology, Second University of Naples Naples, Italy
| |
Collapse
|
34
|
Zeng YY, Jiang WD, Liu Y, Wu P, Zhao J, Jiang J, Kuang SY, Tang L, Tang WN, Zhang YA, Zhou XQ, Feng L. Dietary alpha-linolenic acid/linoleic acid ratios modulate intestinal immunity, tight junctions, anti-oxidant status and mRNA levels of NF-κB p65, MLCK and Nrf2 in juvenile grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2016; 51:351-364. [PMID: 26615102 DOI: 10.1016/j.fsi.2015.11.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 06/05/2023]
Abstract
This study was conducted to investigate the effects of dietary alpha-linolenic acid/linoleic acid (ALA/LNA) ratios on the immune response, tight junctions, antioxidant status and immune-related signaling molecules mRNA levels in the intestine of juvenile grass carp (Ctenopharyngodon idella). A total of 1260 juvenile grass carp with an average initial weight of 8.78 ± 0.03 g were fed diets with different ALA/LNA ratios (0.01, 0.34, 0.68, 1.03, 1.41, 1.76 and 2.15) for 60 days. Results indicated that ALA/LNA ratio of 1.03 significantly increased acid phosphatase, lysozyme activities and complement C3 contents, promoted interleukin 10, transforming growth factor β1 and κB inhibitor α mRNA abundance, whereas suppressed pro-inflammatory cytokines (interleukin 1β, interleukin 8, tumor necrosis factor α and interferon γ2) and signal molecules (IκB kinase β, IκB kinase γ and nuclear factor κB p65) mRNA levels in the intestine (P < 0.05), suggesting that optimal dietary ALA/LNA ratio improved intestinal immune response of juvenile fish. Additionally, ALA/LNA ratio of 1.03 significantly promoted Claudin-3, Claudin-b, Claudin-c, Occludin and ZO-1 gene transcription, whereas reduced Claudin-15a and myosin light-chain kinase mRNA levels in the intestine, suggesting that appropriate dietary ALA/LNA ratio strengthened tight junctions in the intestine of juvenile fish. Meanwhile, ALA/LNA ratio of 1.03 noticeably elevated glutathione contents, copper/zinc superoxide dismutase, glutathione peroxidase, glutathione S-transferase and glutathione reductase activities and mRNA levels, as well as signaling molecule nuclear factor erythoid 2-related factor 2 gene transcriptional abundance in the intestine, suggesting that proper ratio of dietary ALA/LNA ameliorate the intestinal antioxidant status of juvenile fish. Based on the quadratic regression analysis of the complement C3 content in the distal intestine and malondialdehyde content in the whole intestine, optimal ALA/LNA ratio for maximum growth of juvenile grass carp (8.78-72.00 g) were estimated to be 1.13 and 1.12, respectively.
Collapse
Affiliation(s)
- Yun-Yun Zeng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Juan Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Wu-Neng Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
35
|
Cavaliere G, Trinchese G, Bergamo P, De Filippo C, Mattace Raso G, Gifuni G, Putti R, Moni BH, Canani RB, Meli R, Mollica MP. Polyunsaturated Fatty Acids Attenuate Diet Induced Obesity and Insulin Resistance, Modulating Mitochondrial Respiratory Uncoupling in Rat Skeletal Muscle. PLoS One 2016; 11:e0149033. [PMID: 26901315 PMCID: PMC4762694 DOI: 10.1371/journal.pone.0149033] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 01/26/2016] [Indexed: 01/08/2023] Open
Abstract
Objectives Omega (ω)-3 polyunsaturated fatty acids (PUFA) are dietary compounds able to attenuate insulin resistance. Anyway, the precise actions of ω-3PUFAs in skeletal muscle are overlooked. We hypothesized that PUFAs, modulating mitochondrial function and efficiency, would ameliorate pro-inflammatory and pro-oxidant signs of nutritionally induced obesity. Study Design To this aim, rats were fed a control diet (CD) or isocaloric high fat diets containing either ω-3 PUFA (FD) or lard (LD) for 6 weeks. Results FD rats showed lower weight, lipid gain and energy efficiency compared to LD-fed animals, showing higher energy expenditure and O2 consumption/CO2 production. Serum lipid profile and pro-inflammatory parameters in FD-fed animals were reduced compared to LD. Accordingly, FD rats exhibited a higher glucose tolerance revealed by an improved glucose and insulin tolerance tests compared to LD, accompanied by a restoration of insulin signalling in skeletal muscle. PUFAs increased lipid oxidation and reduced energy efficiency in subsarcolemmal mitochondria, and increase AMPK activation, reducing both endoplasmic reticulum and oxidative stress. Increased mitochondrial respiration was related to an increased mitochondriogenesis in FD skeletal muscle, as shown by the increase in PGC1-α and -β. Conclusions our data strengthened the association of high dietary ω3-PUFA intake with reduced mitochondrial energy efficiency in the skeletal muscle.
Collapse
Affiliation(s)
- Gina Cavaliere
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Giovanna Trinchese
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Paolo Bergamo
- Institute of Food Sciences, CNR-ISA, Avellino, Italy
| | - Chiara De Filippo
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | | | - Giorgio Gifuni
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Rosalba Putti
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Bottu Heleena Moni
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Sciences, European Laboratory for Food Induced Diseases, University of Naples "Federico II", Napoli, Italy
| | - Rosaria Meli
- Department of Pharmacy, University of Naples "Federico II", Napoli, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| |
Collapse
|
36
|
Abstract
Mechanistic target of rapamycin (mTOR, also known as mammalian target of rapamycin) is a ubiquitous serine/threonine kinase that regulates cell growth, proliferation and survival. These effects are cell-type-specific, and are elicited in response to stimulation by growth factors, hormones and cytokines, as well as to internal and external metabolic cues. Rapamycin was initially developed as an inhibitor of T-cell proliferation and allograft rejection in the organ transplant setting. Subsequently, its molecular target (mTOR) was identified as a component of two interacting complexes, mTORC1 and mTORC2, that regulate T-cell lineage specification and macrophage differentiation. mTORC1 drives the proinflammatory expansion of T helper (TH) type 1, TH17, and CD4(-)CD8(-) (double-negative, DN) T cells. Both mTORC1 and mTORC2 inhibit the development of CD4(+)CD25(+)FoxP3(+) T regulatory (TREG) cells and, indirectly, mTORC2 favours the expansion of T follicular helper (TFH) cells which, similarly to DN T cells, promote B-cell activation and autoantibody production. In contrast to this proinflammatory effect of mTORC2, mTORC1 favours, to some extent, an anti-inflammatory macrophage polarization that is protective against infections and tissue inflammation. Outside the immune system, mTORC1 controls fibroblast proliferation and chondrocyte survival, with implications for tissue fibrosis and osteoarthritis, respectively. Rapamycin (which primarily inhibits mTORC1), ATP-competitive, dual mTORC1/mTORC2 inhibitors and upstream regulators of the mTOR pathway are being developed to treat autoimmune, hyperproliferative and degenerative diseases. In this regard, mTOR blockade promises to increase life expectancy through treatment and prevention of rheumatic diseases.
Collapse
Affiliation(s)
- Andras Perl
- Division of Rheumatology, Departments of Medicine, Microbiology and Immunology, and Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, College of Medicine, 750 East Adams Street, Syracuse, New York 13210, USA
| |
Collapse
|
37
|
Dietary extra virgin olive oil attenuates kidney injury in pristane-induced SLE model via activation of HO-1/Nrf-2 antioxidant pathway and suppression of JAK/STAT, NF-κB and MAPK activation. J Nutr Biochem 2015; 27:278-88. [PMID: 26525667 DOI: 10.1016/j.jnutbio.2015.09.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 09/14/2015] [Accepted: 09/18/2015] [Indexed: 12/30/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by a widespread organ involvement. Recent studies have suggested that extra virgin olive oil (EVOO) might possess preventive effects on this immunoinflammation-related disease. However, its role in SLE remained unknown. In this work, we evaluated the effects of EVOO diet in a pristane-induced SLE model in mice. Three-month-old mice received an injection of pristane or saline solution and were fed with different experimental diets: sunflower oil diet or EVOO diet. After 24weeks, mice were sacrificed, spleens were collected and kidneys were removed for immunoinflammatory detections. The kidney expression of microsomal prostaglandin E synthase 1, heme oxygenase 1 (HO-1), nuclear factor E2-related factor 2 (Nrf-2), mitogen-activated protein kinases (MAPKs), Janus kinase/signal transducer and activator of transcription (JAK/STAT) and nuclear transcription factor-kappa B (NF-κB) pathways were studied by western blotting. In addition to macroscopic and histological analyses, serum matrix metalloproteinase 3 (MMP-3) levels and proinflammatory cytokines production in splenocytes were evaluated by enzyme-linked immunoassay. We have demonstrated that EVOO diet significantly reduced renal damage and decreased MMP-3 serum and PGE2 kidney levels as well as the proinflammatory cytokines production in splenocytes. Our data indicate that Nrf-2 and HO-1 protein expressions were up-regulated in those mice fed with EVOO and the activation of JAK/STAT, MAPK and NF-κB pathways were drastically ameliorated. These results support the interest of EVOO as a beneficial functional food exerting a preventive/palliative role in the management of SLE.
Collapse
|
38
|
Basiricò L, Morera P, Dipasquale D, Tröscher A, Serra A, Mele M, Bernabucci U. Conjugated linoleic acid isomers strongly improve the redox status of bovine mammary epithelial cells (BME-UV1). J Dairy Sci 2015; 98:7071-82. [PMID: 26277317 DOI: 10.3168/jds.2015-9787] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/18/2015] [Indexed: 12/31/2022]
Abstract
Some studies have shown the protective effects of conjugated linoleic acid (CLA) isomers against oxidative stress and lipid peroxidation in animal models, but no information is available about CLA and changes in oxidative status of the bovine mammary gland. The objectives of the study were to assess in vitro the effect of CLA on the cellular antioxidant response of bovine mammary cells, to examine whether CLA isomers could play a role in cell protection against the oxidative stress, and to study the molecular mechanism involved. For the study, BME-UV1 cells, a bovine mammary epithelial cell line, were used as the experimental model. The BME-UV1 cells were treated with complete medium containing 50 µM cis-9,trans-11 CLA (c9,t11 CLA), trans-10,cis-12 CLA (t10,c12 CLA), and CLA mixture (1:1, cis-9,trans-11: trans-10,cis-12 CLA). To monitor cellular uptake of CLA isomers, cells and culture medium were collected at 0, 3, and 48 h from CLA addition for lipid extraction and fatty acid analyses. To assess the cellular antioxidant response, glutathione (GSH/GSSH), NADPH, and γ-glutamyl-cysteine ligase activity was measured after 48 h from addition of CLA. Cytoplasmic superoxide dismutase, glutathione peroxidase, glutathione S-transferase, and glutathione reductase activities and mRNA were also determined. Intracellular reactive oxygen species and thiobarbituric acid reactive substance production were assessed in cells supplemented with CLA isomers. Cell viability after 3h to H2O2 exposure was assessed to evaluate and to compare the potential protection of different CLA isomers against H2O2-induced oxidative stress. Mammary cells readily picked up all CLA isomers, their accumulation was time dependent, and main metabolites at 48 h are two 18:3 isomers. The CLA treatment induced an intracellular GSH increase, matched by high concentration of NADPH, and an increase of γ-glutamyl-cysteine ligase activity mainly in cells treated with the t10,c12 CLA isomer. The CLA isomer treatment of bovine mammary cells increased superoxide dismutase, glutathione peroxidase, and glutathione S-transferase activity and decreased glutathione reductase activity, but no changes in gene expression of these antioxidant enzymes were observed. Cells supplemented with CLA isomers showed a reduction in intracellular reactive oxygen species and thiobarbituric acid reactive substance levels. All CLA isomers were able to enhance cell resistance against H2O2-induced oxidative stress. These suggest an antioxidant role of CLA, in particular of t10,c12 CLA, by developing a significantly high redox status in cells.
Collapse
Affiliation(s)
- L Basiricò
- Dipartimento di scienze e tecnologie per l'Agricoltura, le Foreste, la Natura e l'Energia, Università degli Studi della Tuscia, 01100, Viterbo, Italy
| | - P Morera
- Dipartimento di scienze e tecnologie per l'Agricoltura, le Foreste, la Natura e l'Energia, Università degli Studi della Tuscia, 01100, Viterbo, Italy
| | - D Dipasquale
- Dipartimento di scienze e tecnologie per l'Agricoltura, le Foreste, la Natura e l'Energia, Università degli Studi della Tuscia, 01100, Viterbo, Italy
| | | | - A Serra
- Dipartimento di Scienze Agrarie, Alimentari e Agro-ambientali, Università di Pisa, 56126, Pisa, Italy
| | - M Mele
- Dipartimento di Scienze Agrarie, Alimentari e Agro-ambientali, Università di Pisa, 56126, Pisa, Italy
| | - U Bernabucci
- Dipartimento di scienze e tecnologie per l'Agricoltura, le Foreste, la Natura e l'Energia, Università degli Studi della Tuscia, 01100, Viterbo, Italy.
| |
Collapse
|
39
|
Trinchese G, Cavaliere G, Canani RB, Matamoros S, Bergamo P, De Filippo C, Aceto S, Gaita M, Cerino P, Negri R, Greco L, Cani PD, Mollica MP. Human, donkey and cow milk differently affects energy efficiency and inflammatory state by modulating mitochondrial function and gut microbiota. J Nutr Biochem 2015; 26:1136-46. [PMID: 26118693 DOI: 10.1016/j.jnutbio.2015.05.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 02/07/2023]
Abstract
Different nutritional components are able, by modulating mitochondrial function and gut microbiota composition, to influence body composition, metabolic homeostasis and inflammatory state. In this study, we aimed to evaluate the effects produced by the supplementation of different milks on energy balance, inflammatory state, oxidative stress and antioxidant/detoxifying enzyme activities and to investigate the role of the mitochondrial efficiency and the gut microbiota in the regulation of metabolic functions in an animal model. We compared the intake of human milk, gold standard for infant nutrition, with equicaloric supplementation of donkey milk, the best substitute for newborns due to its nutritional properties, and cow milk, the primary marketed product. The results showed a hypolipidemic effect produced by donkey and human milk intake in parallel with enhanced mitochondrial activity/proton leakage. Reduced mitochondrial energy efficiency and proinflammatory signals (tumor necrosis factor α, interleukin-1 and lipopolysaccharide levels) were associated with a significant increase of antioxidants (total thiols) and detoxifying enzyme activities (glutathione-S-transferase, NADH quinone oxidoreductase) in donkey- and human milk-treated animals. The beneficial effects were attributable, at least in part, to the activation of the nuclear factor erythroid-2-related factor-2 pathway. Moreover, the metabolic benefits induced by human and donkey milk may be related to the modulation of gut microbiota. In fact, milk treatments uniquely affected the proportions of bacterial phyla and genera, and we hypothesized that the increased concentration of fecal butyrate in human and donkey milk-treated rats was related to the improved lipid and glucose metabolism and detoxifying activities.
Collapse
Affiliation(s)
- Giovanna Trinchese
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Gina Cavaliere
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Sciences, European Laboratory for Food Induced Diseases, Napoli, Italy
| | - Sebastien Matamoros
- Université catholique de Louvain, Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Metabolism and Nutrition research group, Brussels, Belgium
| | - Paolo Bergamo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Chiara De Filippo
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Serena Aceto
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Marcello Gaita
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Pellegrino Cerino
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Rossella Negri
- Department of Translational Medical Sciences, European Laboratory for Food Induced Diseases, Napoli, Italy
| | - Luigi Greco
- Department of Translational Medical Sciences, European Laboratory for Food Induced Diseases, Napoli, Italy
| | - Patrice D Cani
- Université catholique de Louvain, Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Metabolism and Nutrition research group, Brussels, Belgium
| | - Maria Pina Mollica
- Department of Biology, University of Naples "Federico II", Napoli, Italy.
| |
Collapse
|
40
|
Ogita T, Bergamo P, Maurano F, D'Arienzo R, Mazzarella G, Bozzella G, Luongo D, Sashihara T, Suzuki T, Tanabe S, Rossi M. Modulatory activity of Lactobacillus rhamnosus OLL2838 in a mouse model of intestinal immunopathology. Immunobiology 2015; 220:701-710. [PMID: 25623030 DOI: 10.1016/j.imbio.2015.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 01/09/2015] [Accepted: 01/10/2015] [Indexed: 12/29/2022]
Abstract
Gut microbiota and probiotic strains play an important role in oral tolerance by modulating regulatory and effector cell components of the immune system. We have previously described the ability of Lactobacilli to influence both the innate and adaptive immunity to wheat gluten, a food antigen, in mouse. In this study, we further explored the immunomodulatory mechanisms elicited in this model by testing three specific probiotic strains, namely L. rhamnosus OLL2838, B. infantis ATCC15697 and S. thermophilus Sfi39. In vitro analysis showed the all tested strains induced maturation of bone marrow derived dendritic cells (DCs). However, only L. rhamnosus induced appreciable levels of IL-10 and nitric oxide productions, whereas S. thermophilus essentially elicited IL-12 and TNF-α. The anti-inflammatory ability of OLL2838 was then tested in vivo by adopting mice that develop a gluten-specific enteropathy. This model is characterized by villus blunting, crypt hyperplasia, high levels of intestinal IFN-γ, increased cell apoptosis in lamina propria, and reduced intestinal total glutathione (GSHtot) and glutathione S-transferase (GST) activity. We found that, following administration of OLL2838, GSHtot and GST activity were enhanced, whereas caspase-3 activity was reduced. On the contrary, this probiotic strain failed in recovering the normal histology and further increased intestinal IFN-γ. Confocal microscopy revealed the inability of the probiotic strain to appropriately interact with enterocytes of the small intestine and with Peyer's patches in treated mice. In conclusion, these data highlighted the potential of L. rhamnosus OLL2838 to recover specific toxicity parameters induced by gluten in enteropathic mice through mechanisms that involve induction of low levels of reactive oxygen species (ROS).
Collapse
Affiliation(s)
- Tasuku Ogita
- National Agriculture and Food Research Organization, National Food Research Institute Food Function Division, Functional Food Factor Laboratory, Tsukuba, Japan
| | - Paolo Bergamo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Francesco Maurano
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Rossana D'Arienzo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | | | | | - Diomira Luongo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | | | - Takuya Suzuki
- Hiroshima University, Graduate School of Biosphere Science, Hiroshima, Japan
| | - Soichi Tanabe
- Hiroshima University, Graduate School of Biosphere Science, Hiroshima, Japan
| | - Mauro Rossi
- Institute of Food Sciences, National Research Council, Avellino, Italy.
| |
Collapse
|
41
|
Adaptive response activated by dietary cis9, trans11 conjugated linoleic acid prevents distinct signs of gliadin-induced enteropathy in mice. Eur J Nutr 2015; 55:729-740. [PMID: 25840667 DOI: 10.1007/s00394-015-0893-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 03/25/2015] [Indexed: 12/19/2022]
Abstract
PURPOSE The beneficial effects of conjugated linoleic acid (CLA) mixture (cis9, trans11, c9; trans10, cis12, t10) against gliadin-induced toxicity in HLA-DQ8-transgenic mice (DQ8) have been associated with improved duodenal cytoprotective mechanisms [nuclear factor-E2-related factor-2, Nrf2; acylpeptide hydrolase (APEH)/proteasome]. The present study was aimed at investigating the ability of individual CLA isomers to improve the efficacy of these defensive mechanisms and to protect against duodenal injury caused by the combined administration of gliadin and indomethacin (GI). METHODS Gluten-mediated enteropathy was induced in DQ8 mice by three intra-gastric administration of gliadin (20 mg kg(-1)/bw) and indomethacin (15 mg L(-1)) in drinking water for 10 days (GI). C9 or t10 CLA (520 mg kg(-1)/bw/day) were orally administered for 2 weeks. Pro-oxidant and toxic effects associated with GI treatment, anti-oxidant/detoxifying ability of c9 or t10-CLA and the protective effect induced by c9 pre-treatment (c9 + GI) were evaluated in DQ8 mice duodenum by combining enzymatic, immunoblotting, histological evaluation and quantitative real-time PCR assays. RESULTS GI treatment produces the time-dependent decline of the considered detoxifying mechanisms thus leading to pro-apoptotic and pro-oxidant effects. APEH/proteasome pathway was not markedly affected by individual CLA isomers, but duodenal redox status and activity/mRNA levels of Nrf2-activated enzymes were significantly improved by c9 administration. c9 pre-treatment protects against GI-mediated accumulation of oxidative stress markers, and histological examination reveals the increase of goblet cells number in mouse duodenum but induces only a partial recovery of APEH/proteasome activity. CONCLUSIONS The activation of and adaptive response by low doses of c9 supplementation prevents distinct signs of gliadin-induced enteropathy in DQ8 mice.
Collapse
|
42
|
Mazzarella G, Bergamo P, Maurano F, Luongo D, Rotondi Aufiero V, Bozzella G, Palmieri G, Troncone R, Auricchio S, David C, Rossi M. Gliadin intake alters the small intestinal mucosa in indomethacin-treated HLA-DQ8 transgenic mice. Am J Physiol Gastrointest Liver Physiol 2014; 307:G302-G312. [PMID: 24924747 DOI: 10.1152/ajpgi.00002.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Celiac disease (CD) is an enteropathy caused by the ingestion of wheat gluten in genetically susceptible individuals. A complete understanding of the pathogenic mechanisms in CD has been hindered because of the lack of adequate in vivo models. In the present study, we explored the events after the intragastric administration of gliadin and of the albumin/globulin fraction from wheat in human leukocyte antigen-DQ8 transgenic mice (DQ8 mice) treated with indomethacin, an inhibitor of cyclooxygenases (COXs). After 10 days of treatment, mice showed a significant reduction of villus height, increased crypt depth, increased number of lamina propria-activated macrophages, and high basal interferon-γ secretion in mesenteric lymph nodes, all of which were specifically related to gliadin intake, whereas the albumin/globulin fraction of wheat was unable to induce similar changes. Cotreatment with NS-398, a specific inhibitor of COX-2, also induced the intestinal lesion. Enteropathy onset was further characterized by high levels of oxidative stress markers, similar to CD. Biochemical assessment of the small intestine revealed the specific activation of matrix metalloproteinases 2 and 9, high caspase-3 activity, and a significant increase of tissue transglutaminase protein levels associated with the intestinal lesion. Notably, after 30 days of treatment, enteropathic mice developed serum antibodies toward gliadin (IgA) and tissue transglutaminase (IgG). We concluded that gliadin intake in combination with COX inhibition caused a basal inflammatory status and an oxidative stress condition in the small intestine of DQ8 mice, thus triggering the mucosal lesion and, subsequently, an antigen-specific immunity.
Collapse
Affiliation(s)
| | - Paolo Bergamo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Francesco Maurano
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Diomira Luongo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | | | | | - Gianna Palmieri
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Riccardo Troncone
- European Laboratory for Investigation of Food Induced Diseases and Department of Pediatrics, University "Federico II" of Naples, Naples, Italy; and
| | - Salvatore Auricchio
- European Laboratory for Investigation of Food Induced Diseases and Department of Pediatrics, University "Federico II" of Naples, Naples, Italy; and
| | - Chella David
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Mauro Rossi
- Institute of Food Sciences, National Research Council, Avellino, Italy;
| |
Collapse
|
43
|
Mollica MP, Trinchese G, Cavaliere G, De Filippo C, Cocca E, Gaita M, Della-Gatta A, Marano A, Mazzarella G, Bergamo P. c9,t11-Conjugated linoleic acid ameliorates steatosis by modulating mitochondrial uncoupling and Nrf2 pathway. J Lipid Res 2014; 55:837-849. [PMID: 24634500 PMCID: PMC3995462 DOI: 10.1194/jlr.m044032] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 03/14/2014] [Indexed: 12/30/2022] Open
Abstract
Oxidative stress, hepatic steatosis, and mitochondrial dysfunction are key pathophysiological features of nonalcoholic fatty liver disease. A conjugated linoleic acid (CLA) mixture of cis9,trans11 (9,11-CLA) and trans10,cis12 (10,12-CLA) isomers enhanced the antioxidant/detoxifying mechanism via the activation of nuclear factor E2-related factor-2 (Nrf2) and improved mitochondrial function, but less is known about the actions of specific isomers. The differential ability of individual CLA isomers to modulate these pathways was explored in Wistar rats fed for 4 weeks with a lard-based high-fat diet (L) or with control diet (CD), and, within each dietary treatment, two subgroups were daily administered with 9,11-CLA or 10,12-CLA (30 mg/day). The 9,11-CLA, but not 10,12-CLA, supplementation to CD rats improves the GSH/GSSG ratio in the liver, mitochondrial functions, and Nrf2 activity. Histological examination reveals a reduction of steatosis in L-fed rats supplemented with both CLA isomers, but 9,11-CLA downregulated plasma concentrations of proinflammatory markers, mitochondrial dysfunction, and oxidative stress markers in liver more efficiently than in 10,12-CLA treatment. The present study demonstrates the higher protective effect of 9,11-CLA against diet-induced pro-oxidant and proinflammatory signs and suggests that these effects are determined, at least in part, by its ability to activate the Nrf2 pathway and to improve the mitochondrial functioning and biogenesis.
Collapse
Affiliation(s)
- Maria Pina Mollica
- Dipartimento delle Scienze Biologiche, Università degli Studi di Napoli “Federico II,” Napoli, Italy
| | - Giovanna Trinchese
- Dipartimento delle Scienze Biologiche, Università degli Studi di Napoli “Federico II,” Napoli, Italy
| | - Gina Cavaliere
- Dipartimento delle Scienze Biologiche, Università degli Studi di Napoli “Federico II,” Napoli, Italy
| | - Chiara De Filippo
- Dipartimento delle Scienze Biologiche, Università degli Studi di Napoli “Federico II,” Napoli, Italy
| | - Ennio Cocca
- Institute of Biosciences and BioResources, National Research Council (CNR-IBBR), Napoli, Italy
| | - Marcello Gaita
- Dipartimento delle Scienze Biologiche, Università degli Studi di Napoli “Federico II,” Napoli, Italy
| | - Antonio Della-Gatta
- Dipartimento delle Scienze Biologiche, Università degli Studi di Napoli “Federico II,” Napoli, Italy
| | - Angela Marano
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Giuseppe Mazzarella
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Paolo Bergamo
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| |
Collapse
|
44
|
Huebner SM, Olson JM, Campbell JP, Bishop JW, Crump PM, Cook ME. Dietary trans-10,cis-12 CLA reduces murine collagen-induced arthritis in a dose-dependent manner. J Nutr 2014; 144:177-84. [PMID: 24285692 DOI: 10.3945/jn.113.182550] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Dietary trans-10,cis-12 (t10c12) conjugated linoleic acid (CLA) has been shown to reduce inflammation in a murine collagen-induced arthritis (CA) model. To understand the anti-inflammatory potential of t10c12-CLA in the diet, the minimum dose of pure dietary t10c12-CLA capable of reducing CA was investigated. Because plasma inflammatory cytokines often do not reflect the progression of late-stage arthritis, inflamed tissue cytokine concentrations were also investigated in relation to increasing dietary t10c12-CLA amounts. Mice were randomly assigned to the following dietary treatments upon the establishment of arthritis: corn oil (CO) or 0.125%, 0.25%, 0.375%, or 0.5% t10c12-CLA (wt:wt) for 84 d. Sham mice (no arthritis) were fed CO and served as controls. Arthritic paw score, based on subjective assessment of arthritic severity, and paw thickness decreased linearly overall [16-65% (P < 0.001) and 0.5-12% (P < 0.001), respectively] as dietary t10c12-CLA increased (P < 0.001, R(2) < 0.81). Increasing dietary t10c12-CLA was associated with a decrease in plasma interleukin (IL)-1β at days 21 and 42 compared with CO-fed arthritic mice, such that mice fed ≥0.25% t10c12-CLA had IL-1β concentrations that were similar to sham mice. Plasma cytokines returned to sham mice concentrations by day 63 regardless of treatment; however, an arthritis-induced elevation in paw IL-1β decreased linearly as dietary t10c12-CLA concentrations increased at day 84 (P = 0.007, R(2) = 0.92). Similarly, increasing dietary t10c12-CLA linearly decreased paw tumor necrosis factor (TNF)-α (P = 0.05, R(2) = 0.70). In conclusion, ≥0.125% t10c12-CLA dose-dependently reduced inflammation in a murine CA model.
Collapse
|
45
|
Luongo D, Miyamoto J, Bergamo P, Nazzaro F, Baruzzi F, Sashihara T, Tanabe S, Rossi M. Differential modulation of innate immunity in vitro by probiotic strains of Lactobacillus gasseri. BMC Microbiol 2013; 13:298. [PMID: 24365457 PMCID: PMC3879436 DOI: 10.1186/1471-2180-13-298] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 12/18/2013] [Indexed: 12/17/2022] Open
Abstract
Background Probiotics species appear to differentially regulate the intestinal immune response. Moreover, we have shown that different immune-modulatory abilities can be found among probiotic strains belonging to the same species. In this study, we further addressed this issue while studying L. gasseri, a species that induces relevant immune activities in human patients. Results We determined the ability of two strains of L. gasseri, OLL2809 and L13-Ia, to alter cell surface antigen expression, cytokine production and nuclear erythroid 2-related factor 2 (Nrf2)-mediated cytoprotection in murine bone marrow-derived dendritic cells (DCs) and MODE-K cells, which represent an enterocyte model. Differential effects of L. gasseri strains were observed on the expression of surface markers in mature DCs; nevertheless, both strains dramatically induced production of IL-12, TNF-α and IL-10. Distinctive responses to OLL2809 and L13-Ia were also shown in MODE-K cells by analyzing the expression of MHC II molecules and the secretion of IL-6; however, both L. gasseri strains raised intracellular glutathione. Treatment of immature DCs with culture medium from MODE-K monolayers improved cytoprotection and modified the process of DC maturation by down-regulating the expression of co-stimulatory markers and by altering the cytokine profile. Notably, bacteria-conditioned MODE-K cell medium suppressed the expression of the examined cytokines, whereas cytoprotective defenses were significantly enhanced only in DCs exposed to OLL2809-conditioned medium. These effects were essentially mediated by secreted bacterial metabolites. Conclusions We have demonstrated that L. gasseri strains possess distinctive abilities to modulate in vitro DCs and enterocytes. In particular, our results highlight the potential of metabolites secreted by L. gasseri to influence enterocyte-DC crosstalk. Regulation of cellular mechanisms of innate immunity by selected probiotic strains may contribute to the beneficial effects of these bacteria in gut homeostasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mauro Rossi
- Institute of Food Sciences, NRC, Avellino, Italy.
| |
Collapse
|
46
|
Bergamo P, Cocca E, Palumbo R, Gogliettino M, Rossi M, Palmieri G. RedOx status, proteasome and APEH: insights into anticancer mechanisms of t10,c12-conjugated linoleic acid isomer on A375 melanoma cells. PLoS One 2013; 8:e80900. [PMID: 24260504 PMCID: PMC3834215 DOI: 10.1371/journal.pone.0080900] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 10/17/2013] [Indexed: 01/06/2023] Open
Abstract
This study describes the investigation of the efficiency of conjugated linoleic acid (CLA) isomers in reducing cancer cells viability exploring the role of the oxidative stress and acylpeptide hydrolase (APEH)/proteasome mediated pathways on pro-apoptotic activity of the isomer trans10,cis12 (t10,c12)-CLA. The basal activity/expression levels of APEH and proteasome (β-5 subunit) were preliminarily measured in eight cancer cell lines and the functional relationship between these enzymes was clearly demonstrated through their strong positive correlation. t10,c12-CLA efficiently inhibited the activity of APEH and proteasome isoforms in cell-free assays and the negative correlation between cell viability and caspase 3 activity confirmed the pro-apoptotic role of this isomer. Finally, modulatory effects of t10,c12-CLA on cellular redox status (intracellular glutathione, mRNA levels of antioxidant/detoxifying enzymes activated through NF-E2-related factor 2, Nrf2, pathway) and on APEH/β-5 activity/expression levels, were investigated in A375 melanoma cells. Dose- and time-dependent variations of the considered parameters were established and the resulting pro-apoptotic effects were shown to be associated with an alteration of the redox status and a down-regulation of APEH/proteasome pathway. Therefore, our results support the idea that these events are involved in ROS-dependent apoptosis of t10,c12-CLA-treated A375 cells. The combined inhibition, triggered by t10,c12-CLA, via the modulation of APEH/proteasome and Nrf2 pathway for treating melanoma, is suggested as a subject for further in vivo studies.
Collapse
Affiliation(s)
- Paolo Bergamo
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Ennio Cocca
- Institute of Protein Biochemistry, National Research Council (CNR-IBP), Napoli, Italy
| | - Rosanna Palumbo
- Institute of Biostructure and Bioimaging, National Research Council (CNR-IBB), Napoli, Italy
| | - Marta Gogliettino
- Institute of Protein Biochemistry, National Research Council (CNR-IBP), Napoli, Italy
| | - Mose Rossi
- Institute of Protein Biochemistry, National Research Council (CNR-IBP), Napoli, Italy
| | - Gianna Palmieri
- Institute of Protein Biochemistry, National Research Council (CNR-IBP), Napoli, Italy
- * E-mail:
| |
Collapse
|
47
|
Abstract
Oxidative stress is increased in systemic lupus erythematosus (SLE), and it contributes to immune system dysregulation, abnormal activation and processing of cell-death signals, autoantibody production and fatal comorbidities. Mitochondrial dysfunction in T cells promotes the release of highly diffusible inflammatory lipid hydroperoxides, which spread oxidative stress to other intracellular organelles and through the bloodstream. Oxidative modification of self antigens triggers autoimmunity, and the degree of such modification of serum proteins shows striking correlation with disease activity and organ damage in SLE. In T cells from patients with SLE and animal models of the disease, glutathione, the main intracellular antioxidant, is depleted and serine/threonine-protein kinase mTOR undergoes redox-dependent activation. In turn, reversal of glutathione depletion by application of its amino acid precursor, N-acetylcysteine, improves disease activity in lupus-prone mice; pilot studies in patients with SLE have yielded positive results that warrant further research. Blocking mTOR activation in T cells could conceivably provide a well-tolerated and inexpensive alternative approach to B-cell blockade and traditional immunosuppressive treatments. Nevertheless, compartmentalized oxidative stress in self-reactive T cells, B cells and phagocytic cells might serve to limit autoimmunity and its inhibition could be detrimental. Antioxidant therapy might also be useful in ameliorating damage caused by other treatments. This Review thus seeks to critically evaluate the complexity of oxidative stress and its relevance to the pathogenesis and treatment of SLE.
Collapse
|
48
|
Lombardi E, Bergamo P, Maurano F, Bozzella G, Luongo D, Mazzarella G, Rotondi Aufiero V, Iaquinto G, Rossi M. Selective inhibition of the gliadin-specific, cell-mediated immune response by transamidation with microbial transglutaminase. J Leukoc Biol 2013; 93:479-488. [PMID: 23108099 DOI: 10.1189/jlb.0412182] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CD is an immune-mediated enteropathy caused by the ingestion of wheat gluten. The modification of gluten by intestinal tTGase plays a crucial role in CD pathogenesis. In this study, we observed that extensive transamidation of wheat flour with K-C2H5 by mTGase yielded spf and K-gliadins fractions. By Western blot, we found that these modifications were associated with strongly reduced immune cross-reactivity. With the use of DQ8 tg mice as a model of gluten sensitivity, we observed a dramatic reduction in IFNγ production in gliadin-specific spleen cells challenged with spf and K-gliadins in vitro (n=12; median values: 813 vs. 29 and 99; control vs. spf and K-gliadins, P=0.012 for spf, and P=0.003 for K-gliadins). For spf, we also observed an increase in the IL-10/IFNγ protein ratio (n=12; median values: 0.3 vs. 4.7; control vs. spf, P=0.005). In intestinal biopsies from CD patients challenged in vitro with gliadins (n=10), we demonstrated further that K-gliadins dramatically reduced the levels of antigen-specific IFNγ mRNA in all specimens responsive to native gliadins (four of 10; P<0.05). As cytotoxic effects have been described for gliadins, we also studied GST and caspase-3 activities using the enterocytic Caco-2 cell line. We found that neither activities were modified by flour transamidation. Our results indicate that K-C2H5 cross-linking via mTGase specifically affects gliadin immunogenicity, reversing the inducible inflammatory response in models of gluten sensitivity without affecting other aspects of the biological activity of gliadins.
Collapse
|
49
|
Effects of conjugated linoleic acid on growth, non-specific immunity, antioxidant capacity, lipid deposition and related gene expression in juvenile large yellow croaker (Larmichthys crocea) fed soyabean oil-based diets. Br J Nutr 2013; 110:1220-32. [DOI: 10.1017/s0007114513000378] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The effects of conjugated linoleic acid (CLA) on growth performance, non-specific immunity, antioxidant capacity, lipid deposition and related gene expression were investigated in the large yellow croaker (Larmichthys crocea). Fish (7·56 (sem 0·60) g) were fed soyabean oil-based diets with graded levels of CLA (0, 0·42, 0·83, 1·70 %) for 70 d. Quantitative PCR was used to assess the effects of CLA on the transcription of inflammation- and fatty acid oxidation-related genes. Growth in fish fed the diet with 0·42 % CLA was significantly higher. Also, phagocytic index and respiratory burst activity were significantly higher in fish fed the diets containing 0·42 and 0·83 % CLA, respectively. Hepatic total antioxidative capacity and catalase activities increased significantly when CLA increased from 0 to 0·83 %, and then decreased with further increase of CLA. However, hepatic malondialdehyde content decreased significantly as dietary CLA increased. Lipid concentration in the whole body and muscle increased significantly with increasing dietary CLA. Transcription of genes related to inflammation (cyclo-oxygenase-2 and IL-β) in the liver and kidney and fatty acid oxidation (carnitine palmitoyl transferase I and acyl CoA oxidase) in the kidney decreased significantly as dietary CLA increased. PPARα and acyl CoA oxidase expression in the liver decreased significantly as CLA increased from 0·42 to 1·70 %. These results strongly suggest that dietary CLA could significantly affect growth performance, non-specific immunity, antioxidant capacity, lipid deposition and transcription of inflammation- and fatty acid oxidation-related genes of the large yellow croaker. This may contribute to our understanding of the mechanisms related to the physiological effects of dietary CLA in fish.
Collapse
|
50
|
Bergamo P, Gogliettino M, Palmieri G, Cocca E, Maurano F, Stefanile R, Balestrieri M, Mazzarella G, David C, Rossi M. Conjugated linoleic acid protects against gliadin-induced depletion of intestinal defenses. Mol Nutr Food Res 2011; 55 Suppl 2:S248-S256. [PMID: 21954188 DOI: 10.1002/mnfr.201100295] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 07/01/2011] [Accepted: 07/13/2011] [Indexed: 12/22/2022]
Abstract
SCOPE The involvement of oxidative stress in gluten-induced toxicity has been evidenced in vitro and in clinical studies but has never been examined in vivo. We recently demonstrated the protective activity of conjugated linoleic acid (CLA), which functions by the activation of nuclear factor erythroid 2-related factor2 (Nrf2), a key transcription factor for the synthesis of antioxidant and detoxifying enzymes (phase 2). Here, we evaluate the involvement of nuclear factor erythroid 2-related factor2 in gliadin-mediated toxicity in human Caco-2 intestinal cells and in gliadin-sensitive human leukocyte antigen-DQ8 transgenic mice (DQ8) and the protective activity of CLA. METHODS AND RESULTS Gliadin effects in differentiated Caco-2 cells and in DQ8 mice, fed with a gliadin-containing diet with or without CLA supplementation, were evaluated by combining enzymatic, immunochemical, immunohistochemical, and quantitative real-time PCR (qRT-PCR) assays. Gliadin toxicity was accompanied by downregulation of phase 2 and elevates proteasome-acylpeptide hydrolase activities in vitro and in vivo. Notably, gliadin was unable to generate severe oxidative stress extent or pathological consequences in DQ8 mice intestine comparable to those found in celiac patients and the alterations produced were hampered by CLA. CONCLUSION The beneficial effects of CLA against the depletion of crucial intestinal cytoprotective defenses indicates a novel nutritional approach for the treatment of intestinal disease associated with altered redox homeostasis.
Collapse
Affiliation(s)
- Paolo Bergamo
- Istituto di Scienze dell'Alimentazione, Consiglio Nazionale delle Ricerche (CNR-ISA), Avellino, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|