1
|
Mussalo L, Lampinen R, Avesani S, Závodná T, Krejčík Z, Kalapudas J, Penttilä E, Löppönen H, Koivisto AM, Malm T, Topinka J, Giugno R, Jalava P, Kanninen KM. Traffic-related ultrafine particles impair mitochondrial functions in human olfactory mucosa cells - Implications for Alzheimer's disease. Redox Biol 2024; 75:103272. [PMID: 39047637 PMCID: PMC11321383 DOI: 10.1016/j.redox.2024.103272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024] Open
Abstract
Constituents of air pollution, the ultrafine particles (UFP) with a diameter of ≤0.1 μm, are considerably related to traffic emissions. Several studies link air pollution to Alzheimer's disease (AD), yet the exact relationship between the two remains poorly understood. Mitochondria are known targets of environmental toxicants, and their dysfunction is associated with neurodegenerative diseases. The olfactory mucosa (OM), located at the rooftop of the nasal cavity, is directly exposed to the environment and in contact with the brain. Mounting evidence suggests that the UFPs can impact the brain directly through the olfactory tract. By using primary human OM cultures established from nasal biopsies of cognitively healthy controls and individuals diagnosed with AD, we aimed to decipher the effects of traffic-related UFPs on mitochondria. The UFP samples were collected from the exhausts of a modern heavy-duty diesel engine (HDE) without aftertreatment systems, run with renewable diesel (A0) and petroleum diesel (A20), and from an engine of a 2019 model diesel passenger car (DI-E6d) equipped with state-of-the-art aftertreatment devices and run with renewable diesel (Euro6). OM cells were exposed to three different UFPs for 24-h and 72-h, after which cellular processes were assessed on the functional and transcriptomic levels. Our results show that UFPs impair mitochondrial functions in primary human OM cells by hampering oxidative phosphorylation (OXPHOS) and redox balance, and the responses of AD cells differ from cognitively healthy controls. RNA-Seq and IPA® revealed inhibition of OXPHOS and mitochondrial dysfunction in response to UFPs A0 and A20. Functional validation confirmed that A0 and A20 impair cellular respiration, decrease ATP levels, and disturb redox balance by altering NAD and glutathione metabolism, leading to increased ROS and oxidative stress. RNA-Seq and functional assessment revealed the presence of AD-related alterations in human OM cells and that different fuels and engine technologies elicit differential effects.
Collapse
Affiliation(s)
- Laura Mussalo
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Riikka Lampinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Simone Avesani
- Department of Computer Science, University of Verona, 37134, Verona, Italy
| | - Táňa Závodná
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Zdeněk Krejčík
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Juho Kalapudas
- Department of Neurology, Neuro Centre, Kuopio University Hospital, 70210, Kuopio, Finland
| | - Elina Penttilä
- Department of Otorhinolaryngology, University of Eastern Finland and Kuopio University Hospital, 70210, Kuopio, Finland
| | - Heikki Löppönen
- Department of Otorhinolaryngology, University of Eastern Finland and Kuopio University Hospital, 70210, Kuopio, Finland
| | - Anne M Koivisto
- Department of Neurology, Neuro Centre, Kuopio University Hospital, 70210, Kuopio, Finland; Brain Research Unit, Department of Neurology, School of Medicine, University of Eastern Finland, 70210, Kuopio, Finland; Department of Neurology and Geriatrics, Helsinki University Hospital and Neurosciences, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Tarja Malm
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Jan Topinka
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Rosalba Giugno
- Department of Computer Science, University of Verona, 37134, Verona, Italy
| | - Pasi Jalava
- Inhalation Toxicology Laboratory, Department of Environmental and Biological Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Katja M Kanninen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland.
| |
Collapse
|
2
|
Boonpraman N, Yi SS. NADPH oxidase 4 (NOX4) as a biomarker and therapeutic target in neurodegenerative diseases. Neural Regen Res 2024; 19:1961-1966. [PMID: 38227522 DOI: 10.4103/1673-5374.390973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/25/2023] [Indexed: 01/17/2024] Open
Abstract
Diseases like Alzheimer's and Parkinson's diseases are defined by inflammation and the damage neurons undergo due to oxidative stress. A primary reactive oxygen species contributor in the central nervous system, NADPH oxidase 4, is viewed as a potential therapeutic touchstone and indicative marker for these ailments. This in-depth review brings to light distinct features of NADPH oxidase 4, responsible for generating superoxide and hydrogen peroxide, emphasizing its pivotal role in activating glial cells, inciting inflammation, and disturbing neuronal functions. Significantly, malfunctioning astrocytes, forming the majority in the central nervous system, play a part in advancing neurodegenerative diseases, due to their reactive oxygen species and inflammatory factor secretion. Our study reveals that aiming at NADPH oxidase 4 within astrocytes could be a viable treatment pathway to reduce oxidative damage and halt neurodegenerative processes. Adjusting NADPH oxidase 4 activity might influence the neuroinflammatory cytokine levels, including myeloperoxidase and osteopontin, offering better prospects for conditions like Alzheimer's disease and Parkinson's disease. This review sheds light on the role of NADPH oxidase 4 in neural degeneration, emphasizing its drug target potential, and paving the path for novel treatment approaches to combat these severe conditions.
Collapse
Affiliation(s)
- Napissara Boonpraman
- BK21 four Program, Department of Medical Sciences, Soonchunhyang University, Asan, South Korea
| | - Sun Shin Yi
- BK21 four Program, Department of Medical Sciences, Soonchunhyang University, Asan, South Korea
- Department of Biomedical Laboratory Science, Soonchunhyang University, Asan, South Korea
- iConnectome, Co., Ltd., Cheonan, South Korea
| |
Collapse
|
3
|
Kurtz M, Lezón C, Masci I, Boyer P, Brites F, Bonetto J, Bozal C, Álvarez L, Tasat D. Air pollution induces morpho-functional, biochemical and biomechanical vascular dysfunction in undernourished rats. Food Chem Toxicol 2024; 190:114777. [PMID: 38824989 DOI: 10.1016/j.fct.2024.114777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/08/2024] [Accepted: 05/29/2024] [Indexed: 06/04/2024]
Abstract
Air pollution (gases and particulate matter -PM) and child undernutrition are globally recognized stressors with significant consequences. PM and its components breach the respiratory alveolar-capillary barrier, entering the vasculature transporting not only harmful particles and its mediators but, altering vascular paracrine and autocrine functions. The aim of this study was to investigate the effects of Residual Oil Fly Ash (ROFA), on the vasculature of young animals with nutritional growth retardation (NGR). Weanling rats were fed a diet restricted 20% (NGR) compared to ad libitum intake (control-C) for 4 weeks. Rats were intranasally instilled with 1 mg/kg BW of ROFA. After 24h exposure, histological and immunohistochemical, biochemical and contractile response to NA/ACh were evaluated in aortas. ROFA induced changes in the tunica media of the aorta in all groups regarding thickness, muscular cells and expression of Connexin-43. ROFA increased TGF-β1 and decreased eNOs levels and calcium channels in C and NGR animals. An increment in cytokines IL-6 and IL-10 was observed in C, with no changes in NGR. ROFA exposure altered the vascular contractile capacity. In conclusion, ROFA exposure could increase the risk for CVD through the alteration of vascular biochemical parameters, a possible step of the endothelial dysfunction.
Collapse
Affiliation(s)
- Melisa Kurtz
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín- CONICET, Buenos Aires, Argentina.
| | - Christian Lezón
- Cátedra de Fisiología, Facultad de Odontología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ivana Masci
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín- CONICET, Buenos Aires, Argentina
| | - Patricia Boyer
- Cátedra de Fisiología, Facultad de Odontología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fernando Brites
- Laboratorio de Lípidos y Aterosclerosis, Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julián Bonetto
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín- CONICET, Buenos Aires, Argentina
| | - Carola Bozal
- Cátedra de Histología y Embriología, Facultad de Odontología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Laura Álvarez
- Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Deborah Tasat
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín- CONICET, Buenos Aires, Argentina; Cátedra de Histología y Embriología, Facultad de Odontología, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
4
|
Esposito E, Pecorelli A, Ferrara F, Lila MA, Valacchi G. Feeding the Body Through the Skin: Ethosomes and Transethosomes as a New Topical Delivery System for Bioactive Compounds. Annu Rev Food Sci Technol 2024; 15:53-78. [PMID: 38941493 DOI: 10.1146/annurev-food-072023-034528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Because the feeding of our body through the oral route can be associated with many drawbacks due to the degradation of natural molecules during transit in the gastrointestinal tract, a transdermal delivery strategy, usually employed in the pharmaceutical field, can present an effective alternative for delivery of bioactives and nutrients from foods. In this review, the chance to feed the body with nutritive and bioactive molecules from food through transdermal administration is discussed. Various nanotechnological devices employed for topical and transdermal delivery of bioactive compounds are described. In addition, mechanisms underlying their potential use in the delivery of nutritive molecules, as well as their capability to efficaciously reach the dermis and promote systemic distribution, are detailed.
Collapse
Affiliation(s)
- Elisabetta Esposito
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Alessandra Pecorelli
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
| | - Francesca Ferrara
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Mary Ann Lila
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, North Carolina, USA
| | - Giuseppe Valacchi
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
- Department of Animal Science, North Carolina State University, Kannapolis, North Carolina, USA;
- Department of Food and Nutrition, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
5
|
Militello R, Luti S, Gamberi T, Pellegrino A, Modesti A, Modesti PA. Physical Activity and Oxidative Stress in Aging. Antioxidants (Basel) 2024; 13:557. [PMID: 38790662 PMCID: PMC11117672 DOI: 10.3390/antiox13050557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/22/2024] [Accepted: 04/27/2024] [Indexed: 05/26/2024] Open
Abstract
Biological aging, characterized by changes in metabolism and physicochemical properties of cells, has an impact on public health. Environment and lifestyle, including factors like diet and physical activity, seem to play a key role in healthy aging. Several studies have shown that regular physical activity can enhance antioxidant defense mechanisms, including the activity of enzymes such as superoxide dismutase (SOD), catalase, and glutathione peroxidase. However, intense or prolonged exercise can also lead to an increase in reactive oxygen species (ROS) production temporarily, resulting in oxidative stress. This phenomenon is referred to as "exercise-induced oxidative stress". The relationship between physical activity and oxidative stress in aging is complex and depends on various factors such as the type, intensity, duration, and frequency of exercise, as well as individual differences in antioxidant capacity and adaptation to exercise. In this review, we analyzed what is reported by several authors regarding the role of physical activity on oxidative stress in the aging process as well as the role of hormesis and physical exercise as tools for the prevention and treatment of sarcopenia, an aging-related disease. Finally, we reported what has recently been studied in relation to the effect of physical activity and sport on aging in women.
Collapse
Affiliation(s)
- Rosamaria Militello
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (R.M.); (S.L.); (T.G.)
| | - Simone Luti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (R.M.); (S.L.); (T.G.)
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (R.M.); (S.L.); (T.G.)
| | - Alessio Pellegrino
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (A.P.); (P.A.M.)
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (R.M.); (S.L.); (T.G.)
| | - Pietro Amedeo Modesti
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (A.P.); (P.A.M.)
| |
Collapse
|
6
|
Kurtz ML, Orona NS, Lezón C, Defosse VC, Astort F, Maglione GA, Boyer PM, Tasat DR. Decreased immune response in undernourished rats after air pollution exposure. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 107:104400. [PMID: 38408716 DOI: 10.1016/j.etap.2024.104400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 02/28/2024]
Abstract
Children are highly vulnerable subpopulation to malnutrition and air pollution. We investigate, in a rat nutritional growth retardation (NGR) model, the impact of Residual Oil Fly Ash (ROFA) on the lung immune response using in vitro and ex vivo methods. In vitro: Alveolar macrophages (AM) were isolated from Control (C) and NGR animals, cultured and treated with ROFA (1-100 µg/ml) for 24 h. Ex vivo: C and NGR rats were intranasally instilled with ROFA (1 mg/kg BW) or PBS. 24 h post-exposure AM were isolated and cultured. ROFA-treatment increased superoxide anion production and TNFα secretion in C-AM in vitro, though for NGR-AM this response was lower. A similar pattern was observed for TNFα and IL-6 secretion in ex vivo experiments. Regarding the antioxidant response, although NGR-AM showed increased Nrf2, after ROFA instillation an attenuated activation was observed. To conclude, chronic undernutrition altered AM response to ROFA affecting immune responsiveness to air pollutants.
Collapse
Affiliation(s)
- Melisa Lidia Kurtz
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín- CONICET, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | - Nadia Soledad Orona
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín- CONICET, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Christian Lezón
- Cátedra de Fisiología, Facultad de Odontología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Verónica Cecilia Defosse
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Buenos Aires, Argentina; Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Francisco Astort
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín- CONICET, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Guillermo Alberto Maglione
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín- CONICET, Buenos Aires, Argentina
| | - Patricia Mónica Boyer
- Cátedra de Fisiología, Facultad de Odontología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Deborah Ruth Tasat
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín- CONICET, Buenos Aires, Argentina; Cátedra de Histología y Embriología, Facultad de Odontología, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
7
|
Caceres L, Abogunloko T, Malchow S, Ehret F, Merz J, Li X, Sol Mitre L, Magnani N, Tasat D, Mwinyella T, Spiga L, Suchanek D, Fischer L, Gorka O, Colin Gissler M, Hilgendorf I, Stachon P, Rog-Zielinska E, Groß O, Westermann D, Evelson P, Wolf D, Marchini T. Molecular mechanisms underlying NLRP3 inflammasome activation and IL-1β production in air pollution fine particulate matter (PM 2.5)-primed macrophages. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 341:122997. [PMID: 38000727 PMCID: PMC10804998 DOI: 10.1016/j.envpol.2023.122997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023]
Abstract
Exposure to air pollution fine particulate matter (PM2.5) aggravates respiratory and cardiovascular diseases. It has been proposed that PM2.5 uptake by alveolar macrophages promotes local inflammation that ignites a systemic response, but precise underlying mechanisms remain unclear. Here, we demonstrate that PM2.5 phagocytosis leads to NLRP3 inflammasome activation and subsequent release of the pro-inflammatory master cytokine IL-1β. Inflammasome priming and assembly was time- and dose-dependent in inflammasome-reporter THP-1-ASC-GFP cells, and consistent across PM2.5 samples of variable chemical composition. While inflammasome activation was promoted by different PM2.5 surrogates, significant IL-1β release could only be observed after stimulation with transition-metal rich Residual Oil Fly Ash (ROFA) particles. This effect was confirmed in primary human monocyte-derived macrophages and murine bone marrow-derived macrophages (BMDMs), and by confocal imaging of inflammasome-reporter ASC-Citrine BMDMs. IL-1β release by ROFA was dependent on the NLRP3 inflammasome, as indicated by lack of IL-1β production in ROFA-exposed NLRP3-deficient (Nlrp3-/-) BMDMs, and by specific NLRP3 inhibition with the pharmacological compound MCC950. In addition, while ROFA promoted the upregulation of pro-inflammatory gene expression and cytokines release, MCC950 reduced TNF-α, IL-6, and CCL2 production. Furthermore, inhibition of TNF-α with a neutralizing antibody decreased IL-1β release in ROFA-exposed BMDMs. Using electron tomography, ROFA particles were observed inside intracellular vesicles and mitochondria, which showed signs of ultrastructural damage. Mechanistically, we identified lysosomal rupture, K+ efflux, and impaired mitochondrial function as important prerequisites for ROFA-mediated IL-1β release. Interestingly, specific inhibition of superoxide anion production (O2•-) from mitochondrial respiratory Complex I, but not III, blunted IL-1β release in ROFA-exposed BMDMs. Our findings unravel the mechanism by which PM2.5 promotes IL-1β release in macrophages and provide a novel link between innate immune response and exposure to air pollution PM2.5.
Collapse
Affiliation(s)
- Lourdes Caceres
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| | - Tijani Abogunloko
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| | - Sara Malchow
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Fabienne Ehret
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Faculty of Biology, University of Freiburg, 79104, Freiburg im Breisgau, Germany
| | - Julian Merz
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Xiaowei Li
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Lucia Sol Mitre
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| | - Natalia Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| | - Deborah Tasat
- Universidad Nacional de General San Martín, Escuela de Ciencia y Tecnología, B1650, General San Martín, Argentina
| | - Timothy Mwinyella
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Lisa Spiga
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Dymphie Suchanek
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Larissa Fischer
- Faculty of Biology, University of Freiburg, 79104, Freiburg im Breisgau, Germany; Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Mark Colin Gissler
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Peter Stachon
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Eva Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| | - Dennis Wolf
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany.
| | - Timoteo Marchini
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| |
Collapse
|
8
|
Smyth T, Jaspers I. Diesel exhaust particles induce polarization state-dependent functional and transcriptional changes in human monocyte-derived macrophages. Am J Physiol Lung Cell Mol Physiol 2024; 326:L83-L97. [PMID: 38084400 PMCID: PMC11279754 DOI: 10.1152/ajplung.00085.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/30/2023] [Accepted: 11/23/2023] [Indexed: 01/10/2024] Open
Abstract
Macrophage populations exist on a spectrum between the proinflammatory M1 and proresolution M2 states and have demonstrated the ability to reprogram between them after exposure to opposing polarization stimuli. Particulate matter (PM) has been repeatedly linked to worsening morbidity and mortality following respiratory infections and has been demonstrated to modify macrophage function and polarization. The purpose of this study was to determine whether diesel exhaust particles (DEP), a key component of airborne PM, would demonstrate polarization state-dependent effects on human monocyte-derived macrophages (hMDMs) and whether DEP would modify macrophage reprogramming. CD14+CD16- monocytes were isolated from the blood of healthy human volunteers and differentiated into macrophages with macrophage colony-stimulating factor (M-CSF). Resulting macrophages were left unpolarized or polarized into the proresolution M2 state before being exposed to DEP, M1-polarizing conditions (IFN-γ and LPS), or both and tested for phagocytic function, secretory profile, gene expression patterns, and bioenergetic properties. Contrary to previous reports, we observed a mixed M1/M2 phenotype in reprogrammed M2 cells when considering the broader range of functional readouts. In addition, we determined that DEP exposure dampens phagocytic function in all polarization states while modifying bioenergetic properties in M1 macrophages preferentially. Together, these data suggest that DEP exposure of reprogrammed M2 macrophages results in a highly inflammatory, highly energetic subpopulation of macrophages that may contribute to the poor health outcomes following PM exposure during respiratory infections.NEW & NOTEWORTHY We determined that reprogramming M2 macrophages in the presence of diesel exhaust particles (DEP) results in a highly inflammatory mixed M1/M2 phenotype. We also demonstrated that M1 macrophages are particularly vulnerable to particulate matter (PM) exposure as seen by dampened phagocytic function and modified bioenergetics. Our study suggests that PM causes reprogrammed M2 macrophages to become a highly energetic, highly secretory subpopulation of macrophages that may contribute to negative health outcomes observed in humans after PM exposure.
Collapse
Affiliation(s)
- Timothy Smyth
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Ilona Jaspers
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
9
|
Marchini T. Redox and inflammatory mechanisms linking air pollution particulate matter with cardiometabolic derangements. Free Radic Biol Med 2023; 209:320-341. [PMID: 37852544 DOI: 10.1016/j.freeradbiomed.2023.10.396] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/27/2023] [Accepted: 10/15/2023] [Indexed: 10/20/2023]
Abstract
Air pollution is the largest environmental risk factor for disease and premature death. Among the different components that are present in polluted air, fine particulate matter below 2.5 μm in diameter (PM2.5) has been identified as the main hazardous constituent. PM2.5 mainly arises from fossil fuel combustion during power generation, industrial processes, and transportation. Exposure to PM2.5 correlates with enhanced mortality risk from cardiovascular diseases (CVD), such as myocardial infarction and stroke. Over the last decade, it has been increasingly suggested that PM2.5 affects CVD already at the stage of risk factor development. Among the multiple biological mechanisms that have been described, the interplay between oxidative stress and inflammation has been consistently highlighted as one of the main drivers of pulmonary, systemic, and cardiovascular effects of PM2.5 exposure. In this context, PM2.5 uptake by tissue-resident immune cells in the lung promotes oxidative and inflammatory mediators release that alter tissue homeostasis at remote locations. This pathway is central for PM2.5 pathogenesis and might account for the accelerated development of risk factors for CVD, including obesity and diabetes. However, transmission and end-organ mechanisms that explain PM2.5-induced impaired function in metabolic active organs are not completely understood. In this review, the main features of PM2.5 physicochemical characteristics related to PM2.5 ability to induce oxidative stress and inflammation will be presented. Hallmark and recent epidemiological and interventional studies will be summarized and discussed in the context of current air quality guidelines and legislation, knowledge gaps, and inequities. Lastly, mechanistic studies at the intersection between redox metabolism, inflammation, and function will be discussed, with focus on heart and adipose tissue alterations. By offering an integrated analysis of PM2.5-induced effects on cardiometabolic derangements, this review aims to contribute to a better understanding of the pathogenesis and potential interventions of air pollution-related CVD.
Collapse
Affiliation(s)
- Timoteo Marchini
- Vascular Immunology Laboratory, Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), Facultad de Farmacia y Bioquímica, C1113AAD, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Fathieh S, Grieve SM, Negishi K, Figtree GA. Potential Biological Mediators of Myocardial and Vascular Complications of Air Pollution-A State-of-the-Art Review. Heart Lung Circ 2023; 32:26-42. [PMID: 36585310 DOI: 10.1016/j.hlc.2022.11.014] [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: 03/09/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 12/29/2022]
Abstract
Ambient air pollution is recognised globally as a significant contributor to the burden of cardiovascular diseases. The evidence from both human and animal studies supporting the cardiovascular impact of exposure to air pollution has grown substantially, implicating numerous pathophysiological pathways and related signalling mediators. In this review, we summarise the list of activated mediators for each pathway that lead to myocardial and vascular injury in response to air pollutants. We performed a systematic search of multiple databases, including articles between 1990 and Jan 2022, summarising the evidence for activated pathways in response to each significant air pollutant. Particulate matter <2.5 μm (PM2.5) was the most studied pollutant, followed by particulate matter between 2.5 μm-10 μm (PM10), nitrogen dioxide (NO2) and ozone (O3). Key pathogenic pathways that emerged included activation of systemic and local inflammation, oxidative stress, endothelial dysfunction, and autonomic dysfunction. We looked at how potential mediators of each of these pathways were linked to both cardiovascular disease and air pollution and included the overlapping mediators. This review illustrates the complex relationship between air pollution and cardiovascular diseases, and discusses challenges in moving beyond associations, towards understanding causal contributions of specific pathways and markers that may inform us regarding an individual's exposure, response, and likely risk.
Collapse
Affiliation(s)
- Sina Fathieh
- Kolling Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Stuart M Grieve
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Radiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Kazuaki Negishi
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tas, Australia; Department of Cardiology, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan; Sydney Medical School Nepean, Faculty of Medicine and Health, Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Nepean Hospital, Sydney, NSW, Australia
| | - Gemma A Figtree
- Kolling Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia.
| |
Collapse
|
11
|
Biological Sensing of Nitric Oxide in Macrophages and Atherosclerosis Using a Ruthenium-Based Sensor. Biomedicines 2022; 10:biomedicines10081807. [PMID: 36009353 PMCID: PMC9405170 DOI: 10.3390/biomedicines10081807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 12/04/2022] Open
Abstract
Macrophage-derived nitric oxide (NO) plays a critical role in atherosclerosis and presents as a potential biomarker. We assessed the uptake, distribution, and NO detection capacity of an irreversible, ruthenium-based, fluorescent NO sensor (Ru-NO) in macrophages, plasma, and atherosclerotic plaques. In vitro, incubation of Ru-NO with human THP1 monocytes and THP1-PMA macrophages caused robust uptake, detected by Ru-NO fluorescence using mass-cytometry, confocal microscopy, and flow cytometry. THP1-PMA macrophages had higher Ru-NO uptake (+13%, p < 0.05) than THP1 monocytes with increased Ru-NO fluorescence following lipopolysaccharide stimulation (+14%, p < 0.05). In mice, intraperitoneal infusion of Ru-NO found Ru-NO uptake was greater in peritoneal CD11b+F4/80+ macrophages (+61%, p < 0.01) than CD11b+F4/80− monocytes. Infusion of Ru-NO into Apoe−/− mice fed high-cholesterol diet (HCD) revealed Ru-NO fluorescence co-localised with atherosclerotic plaque macrophages. When Ru-NO was added ex vivo to aortic cell suspensions from Apoe−/− mice, macrophage-specific uptake of Ru-NO was demonstrated. Ru-NO was added ex vivo to tail-vein blood samples collected monthly from Apoe−/− mice on HCD or chow. The plasma Ru-NO fluorescence signal was higher in HCD than chow-fed mice after 12 weeks (37.9%, p < 0.05). Finally, Ru-NO was added to plasma from patients (N = 50) following clinically-indicated angiograms. There was lower Ru-NO fluorescence from plasma from patients with myocardial infarction (−30.7%, p < 0.01) than those with stable coronary atherosclerosis. In conclusion, Ru-NO is internalised by macrophages in vitro, ex vivo, and in vivo, can be detected in atherosclerotic plaques, and generates measurable changes in fluorescence in murine and human plasma. Ru-NO displays promising utility as a sensor of atherosclerosis.
Collapse
|
12
|
Lakhdar R, Mumby S, Abubakar-Waziri H, Porter A, Adcock IM, Chung KF. Lung toxicity of particulates and gaseous pollutants using ex-vivo airway epithelial cell culture systems. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 305:119323. [PMID: 35447256 DOI: 10.1016/j.envpol.2022.119323] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/14/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
Air pollution consists of a multi-faceted mix of gases and ambient particulate matter (PM) with diverse organic and non-organic chemical components that contribute to increasing morbidity and mortality worldwide. In particular, epidemiological and clinical studies indicate that respiratory health is adversely affected by exposure to air pollution by both causing and worsening (exacerbating) diseases such as chronic obstructive pulmonary disease (COPD), asthma, interstitial pulmonary fibrosis and lung cancer. The molecular mechanisms of air pollution-induced pulmonary toxicity have been evaluated with regards to different types of PM of various sizes and concentrations with single and multiple exposures over different time periods. These data provide a plausible interrelationship between cellular toxicity and the activation of multiple biological processes including proinflammatory responses, oxidative stress, mitochondrial oxidative damage, autophagy, apoptosis, cell genotoxicity, cellular senescence and epithelial-mesenchymal transition. However, these molecular changes have been studied predominantly in cell lines rather than in primary bronchial or nasal cells from healthy subjects or those isolated from patients with airways disease. In addition, they have been conducted under different cell culture conditions and generally in submerged culture rather than the more relevant air-liquid interface culture and with a variety of air pollutant exposure protocols. Cell types may respond differentially to pollution delivered as an aerosol rather than being bathed in media containing agglomerations of particles. As a result, the actual pathophysiological pathways activated by different PMs in primary cells from the airways of healthy and asthmatic subjects remains unclear. This review summarises the literature on the different methodologies utilised in studying the impact of submicron-sized pollutants on cells derived from the respiratory tract with an emphasis on data obtained from primary human cell. We highlight the critical underlying molecular mechanisms that may be important in driving disease processes in response to air pollution in vivo.
Collapse
Affiliation(s)
- Ramzi Lakhdar
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Sharon Mumby
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Hisham Abubakar-Waziri
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Alexandra Porter
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Ian M Adcock
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Kian Fan Chung
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| |
Collapse
|
13
|
Sauvain JJ, Hemmendinger M, Suárez G, Creze C, Hopf NB, Jouannique V, Debatisse A, Pralong JA, Wild P, Canu IG. Malondialdehyde and anion patterns in exhaled breath condensate among subway workers. Part Fibre Toxicol 2022; 19:16. [PMID: 35216613 PMCID: PMC8876786 DOI: 10.1186/s12989-022-00456-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/14/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Underground transportation systems can contribute to the daily particulates and metal exposures for both commuter and subway workers. The redox and metabolic changes in workers exposed to such metal-rich particles have yet to be characterized. We hypothesize that the distribution of nitrosative/oxidative stress and related metabolic biomarkers in exhaled breath condensate (EBC) are modified depending on exposures. RESULTS Particulate number and size as well as mass concentration and airborne metal content were measured in three groups of nine subway workers (station agents, locomotive operators and security guards). In parallel, pre- and post-shift EBC was collected daily during two consecutive working weeks. In this biological matrix, malondialdehyde, lactate, acetate, propionate, butyrate, formate, pyruvate, the sum of nitrite and nitrate (ΣNOx) and the ratio nitrite/nitrate as well as metals and nanoparticle concentrations was determined. Weekly evolution of the log-transformed selected biomarkers as well as their association with exposure variables was investigated using linear mixed effects models with the participant ID as random effect. The professional activity had a strong influence on the pattern of anions and malondialdehyde in EBC. The daily number concentration and the lung deposited surface area of ultrafine particles was consistently and mainly associated with nitrogen oxides variations during the work-shift, with an inhibitory effect on the ΣNOx. We observed that the particulate matter (PM) mass was associated with a decreasing level of acetate, lactate and ΣNOx during the work-shift, suggestive of a build-up of these anions during the previous night in response to exposures from the previous day. Lactate was moderately and positively associated with some metals and with the sub-micrometer particle concentration in EBC. CONCLUSIONS These results are exploratory but suggest that exposure to subway PM could affect concentrations of nitrogen oxides as well as acetate and lactate in EBC of subway workers. The effect is modulated by the particle size and can correspond to the body's cellular responses under oxidative stress to maintain the redox and/or metabolic homeostasis.
Collapse
Affiliation(s)
- Jean-Jacques Sauvain
- Department of Occupational and Environmental Health, Center for Primary Care and Public Health (Unisanté), University Lausanne, Route de la Corniche 2, 1066, Epalinges, Switzerland.
| | - Maud Hemmendinger
- Department of Occupational and Environmental Health, Center for Primary Care and Public Health (Unisanté), University Lausanne, Route de la Corniche 2, 1066, Epalinges, Switzerland
| | - Guillaume Suárez
- Department of Occupational and Environmental Health, Center for Primary Care and Public Health (Unisanté), University Lausanne, Route de la Corniche 2, 1066, Epalinges, Switzerland
| | - Camille Creze
- Department of Occupational and Environmental Health, Center for Primary Care and Public Health (Unisanté), University Lausanne, Route de la Corniche 2, 1066, Epalinges, Switzerland
| | - Nancy B Hopf
- Department of Occupational and Environmental Health, Center for Primary Care and Public Health (Unisanté), University Lausanne, Route de la Corniche 2, 1066, Epalinges, Switzerland
| | - Valérie Jouannique
- Service Santé-Travail, Autonomous Paris Transport Authority (RATP), 88 Boulevard Sébastopol, 75003, Paris, France
| | - Amélie Debatisse
- Service Santé-Travail, Autonomous Paris Transport Authority (RATP), 88 Boulevard Sébastopol, 75003, Paris, France
| | - Jacques A Pralong
- Division of Pulmonary Diseases, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Rue Gabrielle Perret-Gentil 4, 1205, Geneva, Switzerland
| | - Pascal Wild
- Division of Research Management, National Research and Safety Institute (INRS), Rue du Morvan, CS 60027, 54519, Vandoeuvre Cedex, France
| | - Irina Guseva Canu
- Department of Occupational and Environmental Health, Center for Primary Care and Public Health (Unisanté), University Lausanne, Route de la Corniche 2, 1066, Epalinges, Switzerland
| |
Collapse
|
14
|
Quezada-Maldonado EM, Sánchez-Pérez Y, Chirino YI, García-Cuellar CM. Airborne particulate matter induces oxidative damage, DNA adduct formation and alterations in DNA repair pathways. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 287:117313. [PMID: 34022687 DOI: 10.1016/j.envpol.2021.117313] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/12/2021] [Accepted: 05/02/2021] [Indexed: 06/12/2023]
Abstract
Air pollution, which includes particulate matter (PM), is classified in group 1 as a carcinogen to humans by the International Agency for Research in Cancer. Specifically, PM exposure has been associated with lung cancer in patients living in highly polluted cities. The precise mechanism by which PM is linked to cancer has not been completely described, and the genotoxicity induced by PM exposure plays a relevant role in cell damage. In this review, we aimed to analyze the types of DNA damage and alterations in DNA repair pathways induced by PM exposure, from both epidemiological and toxicological studies, to comprehend the contribution of PM exposure to carcinogenesis. Scientific evidence supports that PM exposure mainly causes oxidative stress by reactive oxygen species (ROS) and the formation of DNA adducts, specifically by polycyclic aromatic hydrocarbons (PAH). PM exposure also induces double-strand breaks (DSBs) and deregulates the expression of some proteins in DNA repair pathways, precisely, base and nucleotide excision repairs and homologous repair. Furthermore, specific polymorphisms of DNA repair genes could lead to an adverse response in subjects exposed to PM. Nevertheless, information about the effects of PM on DNA repair pathways is still limited, and it has not been possible to conclude which pathways are the most affected by exposure to PM or if DNA damage is repaired properly. Therefore, deepening the study of genotoxic damage and alterations of DNA repair pathways is needed for a more precise understanding of the carcinogenic mechanism of PM.
Collapse
Affiliation(s)
- Ericka Marel Quezada-Maldonado
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando No. 22, Tlalpan, CP 14080, CDMX, Mexico; Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Unidad de Posgrado Edificio B, Primer Piso, Ciudad Universitaria, Coyoacán, CP 04510, Ciudad de México, Mexico
| | - Yesennia Sánchez-Pérez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando No. 22, Tlalpan, CP 14080, CDMX, Mexico
| | - Yolanda I Chirino
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Los Reyes Iztacala, Tlalnepantla de Baz, CP 54090, Estado de México, Mexico
| | - Claudia M García-Cuellar
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando No. 22, Tlalpan, CP 14080, CDMX, Mexico.
| |
Collapse
|
15
|
Wang Y, Chen D, Liu Y, Zhang Y, Duan C, Otkur W, Chen H, Liu X, Xia T, Qi H, Piao HL, Liu HX. AQP3-mediated H 2 O 2 uptake inhibits LUAD autophagy by inactivating PTEN. Cancer Sci 2021; 112:3278-3292. [PMID: 34091997 PMCID: PMC8353907 DOI: 10.1111/cas.15008] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/29/2021] [Accepted: 06/03/2021] [Indexed: 01/12/2023] Open
Abstract
It is widely accepted that redox reprogramming participates in malignant transformation of lung adenocarcinoma (LUAD). However, the source of excessive reactive oxygen species (ROS) and the downstream signaling regulatory mechanism are complicated and unintelligible. In the current study, we newly identified the aquaporin 3 (AQP3) as a LUAD oncogenic factor with capacity to transport exogenous hydrogen peroxide (H2 O2 ) and increase intracellular ROS levels. Subsequently, we demonstrated that AQP3 was necessary for the facilitated diffusion of exogenous H2 O2 in LUAD cells and that the AQP3-dependent transport of H2 O2 accelerated cell growth and inhibited rapamycin-induced autophagy. Mechanistically, AQP3-mediated H2 O2 uptake increased intracellular ROS levels to inactivate PTEN and activate the AKT/mTOR pathway to subsequently inhibit autophagy and promote proliferation in LUAD cells. Finally, we suggested that AQP3 depletion retarded subcutaneous tumorigenesis in vivo and simultaneously decreased ROS levels and promoted autophagy. These findings underscore the importance of AQP3-induced oxidative stress in malignant transformation and suggest a therapeutic target for LUAD.
Collapse
Affiliation(s)
- Yawei Wang
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China.,CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Di Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yu Liu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yong Zhang
- Department of Pathology, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Chao Duan
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China.,CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Wuxiyar Otkur
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Huan Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Xiaolong Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Tian Xia
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Huan Qi
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.,Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang, China
| | - Hong-Xu Liu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
16
|
Calabró V, Garcés M, Cáceres L, Magnani ND, Marchini T, Freire A, Vico T, Martinefski M, Vanasco V, Tripodi V, Berra A, Alvarez S, Evelson P. Urban air pollution induces alterations in redox metabolism and mitochondrial dysfunction in mice brain cortex. Arch Biochem Biophys 2021; 704:108875. [PMID: 33891961 DOI: 10.1016/j.abb.2021.108875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/21/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
Previous reports indicate that the central nervous system (CNS) is a target of air pollution, causing tissue damage and functional alterations. Oxidative stress and neuroinflammation have been pointed out as possible mechanisms mediating these effects. The aim of this work was to study the chronic effects of urban air pollution on mice brain cortex, focusing on oxidative stress markers, and mitochondrial function. Male 8-week-old BALB/c mice were exposed to filtered air (FA, control) or urban air (UA) inside whole-body exposure chambers, located in a highly polluted area of Buenos Aires city, for up to 4 weeks. Glutathione levels, assessed as GSH/GSSG ratio, were decreased after 1 and 2 weeks of exposure to UA (45% and 25% respectively vs. FA; p < 0.05). A 38% increase in lipid peroxidation was found after 1 week of UA exposure (p < 0.05). Regarding protein oxidation, carbonyl content was significantly increased at week 2 in UA-exposed mice, compared to FA-group, and an even higher increment was found after 4 weeks of exposure (week 2: 40% p < 0.05, week 4: 54% p < 0.001). NADPH oxidase (NOX) and glutathione peroxidase (GPx) activities were augmented at all the studied time points, while superoxide dismutase (Cu,Zn-SOD cytosolic isoform) and glutathione reductase (GR) activities were increased only after 4 weeks of UA exposure (p < 0.05). The increased NOX activity was accompanied with higher expression levels of NOX2 regulatory subunit p47phox, and NOX4 (p < 0.05). Also, UA mice showed impaired mitochondrial function due to a 50% reduction in O2 consumption in active state respiration (p < 0.05), a 29% decrease in mitochondrial inner membrane potential (p < 0.05), a 65% decrease in ATP production rate (p < 0.01) and a 30% increase in H2O2 production (p < 0.01). Moreover, respiratory complexes I-III and II-III activities were decreased in UA group (30% and 36% respectively vs. FA; p < 0.05). UA exposed mice showed alterations in mitochondrial function, increased oxidant production evidenced by NOX activation, macromolecules damage and the onset of the enzymatic antioxidant system. These data indicate that oxidative stress and impaired mitochondrial function may play a key role in CNS damage mechanisms triggered by air pollution.
Collapse
Affiliation(s)
- Valeria Calabró
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina
| | - Mariana Garcés
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina
| | - Lourdes Cáceres
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina
| | - Natalia D Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina
| | - Timoteo Marchini
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina
| | - Agustina Freire
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina
| | - Tamara Vico
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Fisicoquímica, Argentina
| | - Manuela Martinefski
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Argentina
| | - Virginia Vanasco
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Fisicoquímica, Argentina
| | - Valeria Tripodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Argentina
| | - Alejandro Berra
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Centro de Patología Experimental y Aplicada, Argentina
| | - Silvia Alvarez
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Fisicoquímica, Argentina
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina.
| |
Collapse
|
17
|
Garcés M, Magnani ND, Pecorelli A, Calabró V, Marchini T, Cáceres L, Pambianchi E, Galdoporpora J, Vico T, Salgueiro J, Zubillaga M, Moretton MA, Desimone MF, Alvarez S, Valacchi G, Evelson P. Alterations in oxygen metabolism are associated to lung toxicity triggered by silver nanoparticles exposure. Free Radic Biol Med 2021; 166:324-336. [PMID: 33596456 DOI: 10.1016/j.freeradbiomed.2021.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/18/2021] [Accepted: 02/04/2021] [Indexed: 10/22/2022]
Abstract
Along with the AgNP applications development, the concern about their possible toxicity has increasingly gained attention. As the respiratory system is one of the main exposure routes, the aim of this study was to evaluate the harmful effects developed in the lung after an acute AgNP exposure. In vivo studies using Balb/c mice intranasally instilled with 0.1 mg AgNP/kg b.w, were performed. 99mTc-AgNP showed the lung as the main organ of deposition, where, in turn, AgNP may exert barrier injury observed by increased protein content and total cell count in BAL samples. In vivo acute exposure showed altered lung tissue O2 consumption due to increased mitochondrial active respiration and NOX activity. Both O2 consumption processes release ROS triggering the antioxidant system as observed by the increased SOD, catalase and GPx activities and a decreased GSH/GSSG ratio. In addition, increased protein oxidation was observed after AgNP exposure. In A549 cells, exposure to 2.5 μg/mL AgNP during 1 h resulted in augment NOX activity, decreased mitochondrial ATP associated respiration and higher H2O2 production rate. Lung 3D tissue model showed AgNP-initiated barrier alterations as TEER values decreased and morphological alterations. Taken together, these results show that AgNP exposure alters O2 metabolism leading to alterations in oxygen metabolism lung toxicity. AgNP-triggered oxidative damage may be responsible for the impaired lung function observed due to alveolar epithelial injury.
Collapse
Affiliation(s)
- Mariana Garcés
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina
| | - Natalia D Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina
| | - Alessandra Pecorelli
- NC State University, Plants for Human Health Institute, Animal Science Department, USA
| | - Valeria Calabró
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina
| | - Timoteo Marchini
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina
| | - Lourdes Cáceres
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina
| | - Erika Pambianchi
- NC State University, Plants for Human Health Institute, Animal Science Department, USA
| | - Juan Galdoporpora
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química Analítica Instrumental, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Argentina
| | - Tamara Vico
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Fisicoquímica, Argentina
| | - Jimena Salgueiro
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Fisicomatemática, Cátedra de Física, Argentina
| | - Marcela Zubillaga
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Fisicomatemática, Cátedra de Física, Argentina
| | - Marcela A Moretton
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Cátedra de Tecnología Farmacéutica I, Buenos Aires, Argentina
| | - Martin F Desimone
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química Analítica Instrumental, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Argentina
| | - Silvia Alvarez
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Fisicoquímica, Argentina
| | - Giuseppe Valacchi
- NC State University, Plants for Human Health Institute, Animal Science Department, USA; Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy; Kyung Hee University, Department of Food and Nutrition, Seoul, South Korea
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Argentina; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Argentina.
| |
Collapse
|
18
|
Bachmann MC, Bellalta S, Basoalto R, Gómez-Valenzuela F, Jalil Y, Lépez M, Matamoros A, von Bernhardi R. The Challenge by Multiple Environmental and Biological Factors Induce Inflammation in Aging: Their Role in the Promotion of Chronic Disease. Front Immunol 2020; 11:570083. [PMID: 33162985 PMCID: PMC7591463 DOI: 10.3389/fimmu.2020.570083] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
The aging process is driven by multiple mechanisms that lead to changes in energy production, oxidative stress, homeostatic dysregulation and eventually to loss of functionality and increased disease susceptibility. Most aged individuals develop chronic low-grade inflammation, which is an important risk factor for morbidity, physical and cognitive impairment, frailty, and death. At any age, chronic inflammatory diseases are major causes of morbimortality, affecting up to 5-8% of the population of industrialized countries. Several environmental factors can play an important role for modifying the inflammatory state. Genetics accounts for only a small fraction of chronic-inflammatory diseases, whereas environmental factors appear to participate, either with a causative or a promotional role in 50% to 75% of patients. Several of those changes depend on epigenetic changes that will further modify the individual response to additional stimuli. The interaction between inflammation and the environment offers important insights on aging and health. These conditions, often depending on the individual's sex, appear to lead to decreased longevity and physical and cognitive decline. In addition to biological factors, the environment is also involved in the generation of psychological and social context leading to stress. Poor psychological environments and other sources of stress also result in increased inflammation. However, the mechanisms underlying the role of environmental and psychosocial factors and nutrition on the regulation of inflammation, and how the response elicited for those factors interact among them, are poorly understood. Whereas certain deleterious environmental factors result in the generation of oxidative stress driven by an increased production of reactive oxygen and nitrogen species, endoplasmic reticulum stress, and inflammation, other factors, including nutrition (polyunsaturated fatty acids) and behavioral factors (exercise) confer protection against inflammation, oxidative and endoplasmic reticulum stress, and thus ameliorate their deleterious effect. Here, we discuss processes and mechanisms of inflammation associated with environmental factors and behavior, their links to sex and gender, and their overall impact on aging.
Collapse
Affiliation(s)
| | - Sofía Bellalta
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roque Basoalto
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Yorschua Jalil
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Macarena Lépez
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Anibal Matamoros
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Institute of Biological Sciences (ICB), Federal University of Pará, Belem, Brazil
| | - Rommy von Bernhardi
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|