1
|
Li X, Chen K, Liu R, Zheng Z, Hou X. Antimicrobial neuropeptides and their therapeutic potential in vertebrate brain infectious disease. Front Immunol 2024; 15:1496147. [PMID: 39620214 PMCID: PMC11604648 DOI: 10.3389/fimmu.2024.1496147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024] Open
Abstract
The defense mechanisms of the vertebrate brain against infections are at the forefront of immunological studies. Unlike other body parts, the brain not only fends off pathogenic infections but also minimizes the risk of self-damage from immune cell induced inflammation. Some neuropeptides produced by either nerve or immune cells share remarkable similarities with antimicrobial peptides (AMPs) in terms of size, structure, amino acid composition, amphiphilicity, and net cationic charge. These similarities extend to a wide range of antibacterial activities demonstrated in vitro, effectively protecting nerve tissue from microbial threats. This review systematically examines 12 neuropeptides, pituitary adenylate cyclase-activating peptide (PACAP), vasoactive intestinal peptide (VIP), α-melanocyte stimulating hormone (α-MSH), orexin-B (ORXB), ghrelin, substance P (SP), adrenomedullin (AM), calcitonin-gene related peptide (CGRP), urocortin-II (UCN II), neuropeptide Y (NPY), NDA-1, and catestatin (CST), identified for their antimicrobial properties, summarizing their structural features, antimicrobial effectiveness, and action mechanisms. Importantly, the majority of these antimicrobial neuropeptides (9 out of 12) also possess significant anti-inflammatory properties, potentially playing a key role in preserving immune tolerance in various disorders. However, the connection between this anti-inflammatory property and the brain's infection defense strategy has rarely been explored. Our review suggests that the combined antimicrobial and anti-inflammatory actions of neuropeptides could be integral to the brain's defense strategy against pathogens, marking an exciting direction for future research.
Collapse
Affiliation(s)
- Xiaoke Li
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, China
| | - Kaiqi Chen
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, China
| | - Ruonan Liu
- College of Medical Engineering, Jining Medical University, Jining, China
| | - Zhaodi Zheng
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, China
| | - Xitan Hou
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, China
| |
Collapse
|
2
|
Duarte M, Pedrosa SS, Khusial PR, Madureira AR. Exploring the interplay between stress mediators and skin microbiota in shaping age-related hallmarks: A review. Mech Ageing Dev 2024; 220:111956. [PMID: 38906383 DOI: 10.1016/j.mad.2024.111956] [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: 02/16/2024] [Revised: 05/27/2024] [Accepted: 06/14/2024] [Indexed: 06/23/2024]
Abstract
Psychological stress is a major contributing factor to several health problems (e.g., depression, cardiovascular disease). Around 35 % of the world's population suffers from it, including younger generations. Physiologically, stress manifests through neuroendocrine pathways (Hypothalamic-Pituitary-Adrenal (HPA) axis and Sympathetic-Adrenal-Medullary (SAM) system) which culminate in the production of stress mediators like cortisol, epinephrine and norepinephrine. Stress and its mediators have been associated to body aging, through molecular mechanisms such as telomere attrition, mitochondrial dysfunction, cellular senescence, chronic inflammation, and dysbiosis, among others. Regarding its impact in the skin, stress impacts its structural integrity and physiological function. Despite this review focusing on several hallmarks of aging, emphasis was placed on skin microbiota dysbiosis. In this line, several studies, comprising different age groups, demographic contexts and body sites, have reported skin microbiota alterations associated with aging, and some effects of stress mediators on skin microbiota have also been reviewed in this paper. From a different perspective, since it is not a "traditional" stress mediator, oxytocin, a cortisol antagonist, has been related to glucorticoids inhibition and to display positive effects on cellular aging. This hormone dysregulation has been associated to psychological issues such as depression, whereas its upregulation has been linked to positive social interaction.
Collapse
Affiliation(s)
- Marco Duarte
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto 4169-005, Portugal
| | - Sílvia Santos Pedrosa
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto 4169-005, Portugal
| | - P Raaj Khusial
- Amyris Biotech INC, 5885 Hollis St Ste 100, Emeryville, CA 94608-2405, USA
| | - Ana Raquel Madureira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto 4169-005, Portugal.
| |
Collapse
|
3
|
Han JH, Kim HS. Skin Deep: The Potential of Microbiome Cosmetics. J Microbiol 2024; 62:181-199. [PMID: 38625646 DOI: 10.1007/s12275-024-00128-x] [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: 01/02/2024] [Revised: 02/27/2024] [Accepted: 03/03/2024] [Indexed: 04/17/2024]
Abstract
The interplay between the skin microbiome and its host is a complex facet of dermatological health and has become a critical focus in the development of microbiome cosmetics. The skin microbiome, comprising various microorganisms, is essential from birth, develops over the lifespan, and performs vital roles in protecting our body against pathogens, training the immune system, and facilitating the breakdown of organic matter. Dysbiosis, an imbalance of these microorganisms, has been implicated in a number of skin conditions such as acne, atopic dermatitis, and skin cancer. Recent scientific findings have spurred cosmetic companies to develop products that preserve and enhance the skin's microbial diversity balance. These products may incorporate elements like prebiotics, probiotics, and postbiotics, which are beneficial for the skin microbiome. Beyond topical products, there's increasing interest in ingestible beauty supplements (i.e. oral probiotics), highlighting the connection between the gut and skin. This review examines the influence of the microbiome on skin health and the emerging trends of microbiome skincare products.
Collapse
Affiliation(s)
- Ju Hee Han
- Department of Dermatology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Hei Sung Kim
- Department of Dermatology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
4
|
Çetinarslan T, Kümper L, Fölster-Holst R. The immunological and structural epidermal barrier dysfunction and skin microbiome in atopic dermatitis-an update. Front Mol Biosci 2023; 10:1159404. [PMID: 37654796 PMCID: PMC10467310 DOI: 10.3389/fmolb.2023.1159404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 08/04/2023] [Indexed: 09/02/2023] Open
Abstract
Atopic dermatitis (AD) is a common, chronic and relapsing inflammatory skin disease with various clinical presentations and combinations of symptoms. The pathophysiology of AD is complex and multifactorial. There are several factors involved in the etiopathogenesis of AD including structural and immunological epidermal barrier defect, imbalance of the skin microbiome, genetic background and environmental factors. Alterations in structural proteins, lipids, proteases, and their inhibitors, lead to the impairment of the stratum corneum which is associated with the increased skin penetration and transepidermal water loss. The elevated serum immunoglobulin E levels and blood eosinophilia have been shown in the majority of AD patients. Type 2 T-helper cell immune pathway with increased expression of interleukin (IL)-4, IL-5, and IL-13, has an important role in the etiopathogenesis of AD. Both T cells and keratinocytes contribute to epidermal barrier impairment in AD via a dynamic interaction of cytokines and chemokines. The skin microbiome is another factor of relevance in the etiopathogenesis of AD. It has been shown that during AD flares, Staphylococcus aureus (S. aureus) colonization increased, while Staphylococcus epidermidis (S. epidermidis) decreased. On the contrary, S. epidermidis and species of Streptococcus, Corynebacterium and Propionibacterium increased during the remision phases. However, it is not clear whether skin dysbiosis is one of the symptoms or one of the causes of AD. There are several therapeutic options, targeting these pathways which play a critical role in the etiopathogenesis of AD. Although topical steroids are the mainstay of the treatment of AD, new biological therapies including IL-4, IL-13, and IL-31 inhibitors, as well as Janus kinase inhibitors (JAKi), increasingly gain more importance with new advances in the therapy of AD. In this review, we summarize the role of immunological and structural epidermal barrier dysfunction, immune abnormalities, impairment of lipids, filaggrin mutation and skin microbiome in the etiopathogenesis of AD, as well as the therapeutic options for AD and their effects on these abnormalities in AD skin.
Collapse
Affiliation(s)
- Tubanur Çetinarslan
- Department of Dermatology and Venereology, Manisa Celal Bayar University, Manisa, Türkiye
| | - Lisa Kümper
- MEDICE Arzneimittel Pütter GmbH and Co. KG, Iserlohn, Germany
| | - Regina Fölster-Holst
- Department of Dermatology-Venereology and Allergology, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
5
|
Chen Y, Xiao X, Huang C, Zhu J, Zhou H, Qin H, Bao Y, Zhuang T, Zhang G. Flupirtine and antihistamines exert synergistic anti-nociceptive effects in mice. Psychopharmacology (Berl) 2023; 240:881-897. [PMID: 36752814 DOI: 10.1007/s00213-023-06329-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/26/2023] [Indexed: 02/09/2023]
Abstract
RATIONALE Drug combinations are commonly used in pain management, which can produce potent analgesic effects with reduced dosage and adverse effects. OBJECTIVE This study was designed to evaluate the anti-nociceptive effects and adverse effects of new combinations of flupirtine (a Kv7 potassium channel opener) and antihistamines (promethazine, fexofenadine) on acute and chronic pain in mice, and the possible mechanisms behind the synergistic analgesic effects were preliminarily investigated. METHODS In acetic acid writhing test, carrageenan-induced inflammatory pain model, and paclitaxel-induced neuropathic pain model, the interaction indexes (γ) between flupirtine and antihistamines were determined by isobolographic analysis. Furthermore, the Kv7 channel blocker XE991 was used to determine whether the effects of single agents and drug combinations on paclitaxel- and carrageenan-induced mechanical allodynia were mediated by Kv7 channels. Finally, hepatotoxicity markers, liver histopathology, and the rotarod test were used to investigate the adverse effects of drugs in combination doses. RESULTS The interaction indexes of flupirtine-promethazine and flupirtine-fexofenadine in all the above three pain models were lower than 1. The analgesic effects of flupirtine (13 mg/kg), promethazine (5 mg/kg), fexofenadine (20 mg/kg), and their combinations were antagonized significantly by XE991 (3 mg/kg). And the adverse effects of flupirtine and antihistamines in combination doses were not significantly different from the vehicle group. CONCLUSIONS Flupirtine and antihistamines produced synergistic analgesic effects in all the above pain models. The analgesic effects of antihistamines were partially mediated by Kv7/M channels, and the activation of Kv7/M channels may be partly responsible for the synergistic analgesic effects between flupirtine and antihistamines.
Collapse
Affiliation(s)
- Yanming Chen
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xinyi Xiao
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Chaonan Huang
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jin Zhu
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Huiling Zhou
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Huimin Qin
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yu Bao
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Tao Zhuang
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China. .,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Guisen Zhang
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China. .,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
6
|
An Altered Skin and Gut Microbiota Are Involved in the Modulation of Itch in Atopic Dermatitis. Cells 2022; 11:cells11233930. [PMID: 36497188 PMCID: PMC9736894 DOI: 10.3390/cells11233930] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/14/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Skin and gut microbiota play an important role in the pathogenesis of atopic dermatitis (AD). An alteration of the microbiota diversity modulates the development and course of AD, e.g., decreased microbiome diversity correlates with disease severity, particularly in lesional skin of AD. Itch is a hallmark of AD with unsatisfying treatment until now. Recent evidence suggests a possible role of microbiota in altering itch in AD through gut-skin-brain interactions. The microbial metabolites, proinflammatory cytokines, and impaired immune response lead to a modulation of histamine-independent itch, disruption of epidermal barrier, and central sensitization of itch mechanisms. The positive impact of probiotics in alleviating itch in AD supports this hypothesis, which may lead to novel strategies for managing itchy skin in AD patients. This review summarizes the emerging findings on the correlation between an altered microbiota and gut-skin-brain axis in AD, especially in modulating itchy skin.
Collapse
|
7
|
Chen H, Zhao Q, Zhong Q, Duan C, Krutmann J, Wang J, Xia J. Skin Microbiome, Metabolome and Skin Phenome, from the Perspectives of Skin as an Ecosystem. PHENOMICS (CHAM, SWITZERLAND) 2022; 2:363-382. [PMID: 36939800 PMCID: PMC9712873 DOI: 10.1007/s43657-022-00073-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/06/2022] [Accepted: 08/11/2022] [Indexed: 11/07/2022]
Abstract
Skin is a complex ecosystem colonized by millions of microorganisms, including bacteria, fungi, and viruses. Skin microbiota is believed to exert critical functions in maintaining host skin health. Profiling the structure of skin microbial community is the first step to overview the ecosystem. However, the community composition is highly individualized and extremely complex. To explore the fundamental factors driving the complexity of the ecosystem, namely the selection pressures, we review the present studies on skin microbiome from the perspectives of ecology. This review summarizes the following: (1) the composition of substances/nutrients in the cutaneous ecological environment that are derived from the host and the environment, highlighting their proposed function on skin microbiota; (2) the features of dominant skin commensals to occupy ecological niches, through self-adaptation and microbe-microbe interactions; (3) how skin microbes, by their structures or bioactive molecules, reshape host skin phenotypes, including skin immunity, maintenance of skin physiology such as pH and hydration, ultraviolet (UV) protection, odor production, and wound healing. This review aims to re-examine the host-microbe interactions from the ecological perspectives and hopefully to give new inspiration to this field.
Collapse
Affiliation(s)
- Huizhen Chen
- grid.8547.e0000 0001 0125 2443Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Qi Zhao
- grid.27255.370000 0004 1761 1174Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012 China
- grid.435557.50000 0004 0518 6318IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, D-40225 Germany
| | - Qian Zhong
- grid.8547.e0000 0001 0125 2443Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Cheng Duan
- grid.8547.e0000 0001 0125 2443Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou, 511458 China
| | - Jean Krutmann
- grid.435557.50000 0004 0518 6318IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, D-40225 Germany
| | - Jiucun Wang
- grid.8547.e0000 0001 0125 2443Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, 200438 China
- grid.506261.60000 0001 0706 7839Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai, 200438 China
| | - Jingjing Xia
- grid.8547.e0000 0001 0125 2443Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou, 511458 China
| |
Collapse
|
8
|
Slominski AT, Slominski RM, Raman C, Chen JY, Athar M, Elmets C. Neuroendocrine signaling in the skin with a special focus on the epidermal neuropeptides. Am J Physiol Cell Physiol 2022; 323:C1757-C1776. [PMID: 36317800 PMCID: PMC9744652 DOI: 10.1152/ajpcell.00147.2022] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 11/07/2022]
Abstract
The skin, which is comprised of the epidermis, dermis, and subcutaneous tissue, is the largest organ in the human body and it plays a crucial role in the regulation of the body's homeostasis. These functions are regulated by local neuroendocrine and immune systems with a plethora of signaling molecules produced by resident and immune cells. In addition, neurotransmitters, endocrine factors, neuropeptides, and cytokines released from nerve endings play a central role in the skin's responses to stress. These molecules act on the corresponding receptors in an intra-, juxta-, para-, or autocrine fashion. The epidermis as the outer most component of skin forms a barrier directly protecting against environmental stressors. This protection is assured by an intrinsic keratinocyte differentiation program, pigmentary system, and local nervous, immune, endocrine, and microbiome elements. These constituents communicate cross-functionally among themselves and with corresponding systems in the dermis and hypodermis to secure the basic epidermal functions to maintain local (skin) and global (systemic) homeostasis. The neurohormonal mediators and cytokines used in these communications regulate physiological skin functions separately or in concert. Disturbances in the functions in these systems lead to cutaneous pathology that includes inflammatory (i.e., psoriasis, allergic, or atopic dermatitis, etc.) and keratinocytic hyperproliferative disorders (i.e., seborrheic and solar keratoses), dysfunction of adnexal structure (i.e., hair follicles, eccrine, and sebaceous glands), hypersensitivity reactions, pigmentary disorders (vitiligo, melasma, and hypo- or hyperpigmentary responses), premature aging, and malignancies (melanoma and nonmelanoma skin cancers). These cellular, molecular, and neural components preserve skin integrity and protect against skin pathologies and can act as "messengers of the skin" to the central organs, all to preserve organismal survival.
Collapse
Affiliation(s)
- Andrzej T Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, Alabama
- VA Medical Center, Birmingham, Alabama
| | - Radomir M Slominski
- Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, Alabama
| | - Chander Raman
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jake Y Chen
- Informatics Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
- VA Medical Center, Birmingham, Alabama
| | - Craig Elmets
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, Alabama
- VA Medical Center, Birmingham, Alabama
| |
Collapse
|
9
|
Cotton and Flax Textiles Leachables Impact Differently Cutaneous Staphylococcus aureus and Staphylococcus epidermidis Biofilm Formation and Cytotoxicity. Life (Basel) 2022; 12:life12040535. [PMID: 35455029 PMCID: PMC9032481 DOI: 10.3390/life12040535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 11/17/2022] Open
Abstract
Bacteria can bind on clothes, but the impacts of textiles leachables on cutaneous bacteria remain unknown. Here, we studied for the first time the effects of cotton and flax obtained through classical and soft ecological agriculture on the representatives S. aureus and S. epidermidis bacteria of the cutaneous microbiota. Crude flax showed an inhibitory potential on S. epidermidis bacterial lawns whereas cotton had no effect. Textile fiber leachables were produced in bacterial culture media, and these extracts were tested on S. aureus and S. epidermidis. Bacterial growth was not impacted, but investigation by the crystal violet technique and confocal microscopy showed that all extracts affected biofilm formation by the two staphylococci species. An influence of cotton and flax culture conditions was clearly observed. Flax extracts had strong inhibitory impacts and induced the formation of mushroom-like defense structures by S. aureus. Conversely, production of biosurfactant by bacteria and their surface properties were not modified. Resistance to antibiotics also remained unchanged. All textile extracts, and particularly soft organic flax, showed strong inhibitory effects on S. aureus and S. epidermidis cytotoxicity on HaCaT keratinocytes. Analysis of flax leachables showed the presence of benzyl alcohol that could partly explain the effects of flax extracts.
Collapse
|
10
|
Raju SV, Sarkar P, Pasupuleti M, Abbasi AM, Al-Farraj DA, Elshikh MS, Elumalai P, Harikrishnan R, Rahman MA, Arockiaraj J. Antibacterial Activity of RM12, a Tachykinin Derivative, Against Pseudomonas aeruginosa. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10274-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
11
|
Fournière M, Bedoux G, Souak D, Bourgougnon N, Feuilloley MGJ, Latire T. Effects of Ulva sp. Extracts on the Growth, Biofilm Production, and Virulence of Skin Bacteria Microbiota: Staphylococcus aureus, Staphylococcus epidermidis, and Cutibacterium acnes Strains. Molecules 2021; 26:4763. [PMID: 34443349 PMCID: PMC8401615 DOI: 10.3390/molecules26164763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/01/2022] Open
Abstract
Ulva sp. is known to be a source of bioactive compounds such as ulvans, but to date, their biological activity on skin commensal and/or opportunistic pathogen bacteria has not been reported. In this study, the effects of poly- and oligosaccharide fractions produced by enzyme-assisted extraction and depolymerization were investigated, for the first time in vitro, on cutaneous bacteria: Staphylococcus aureus, Staphylococcus epidermidis, and Cutibacterium acnes. At 1000 μg/mL, poly- and oligosaccharide fractions did not affect the growth of the bacteria regarding their generation time. Polysaccharide Ulva sp. fractions at 1000 μg/mL did not alter the bacterial biofilm formation, while oligosaccharide fractions modified S. epidermidis and C. acnes biofilm structures. None of the fractions at 1000 μg/mL significantly modified the cytotoxic potential of S. epidermidis and S. aureus towards keratinocytes. However, poly- and oligosaccharide fractions at 1000 μg/mL induced a decrease in the inflammatory potential of both acneic and non-acneic C. acnes strains on keratinocytes of up to 39.8%; the strongest and most significant effect occurred when the bacteria were grown in the presence of polysaccharide fractions. Our research shows that poly- and oligosaccharide Ulva sp. fractions present notable biological activities on cutaneous bacteria, especially towards C. acnes acneic and non-acneic strains, which supports their potential use for dermo-cosmetic applications.
Collapse
Affiliation(s)
- Mathilde Fournière
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Bretagne Sud, 56000 Vannes, France; (G.B.); (N.B.); (T.L.)
- Université Catholique de l’Ouest Bretagne Nord, 22200 Guingamp, France
| | - Gilles Bedoux
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Bretagne Sud, 56000 Vannes, France; (G.B.); (N.B.); (T.L.)
| | - Djouhar Souak
- Laboratoire de Microbiologie Signaux et Microenvironnement LMSM EA4312, Université de Rouen Normandie, 27000 Évreux, France; (D.S.); (M.G.J.F.)
| | - Nathalie Bourgougnon
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Bretagne Sud, 56000 Vannes, France; (G.B.); (N.B.); (T.L.)
| | - Marc G. J. Feuilloley
- Laboratoire de Microbiologie Signaux et Microenvironnement LMSM EA4312, Université de Rouen Normandie, 27000 Évreux, France; (D.S.); (M.G.J.F.)
| | - Thomas Latire
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Bretagne Sud, 56000 Vannes, France; (G.B.); (N.B.); (T.L.)
- Université Catholique de l’Ouest Bretagne Nord, 22200 Guingamp, France
| |
Collapse
|
12
|
Blicharz L, Rudnicka L, Czuwara J, Waśkiel-Burnat A, Goldust M, Olszewska M, Samochocki Z. The Influence of Microbiome Dysbiosis and Bacterial Biofilms on Epidermal Barrier Function in Atopic Dermatitis-An Update. Int J Mol Sci 2021; 22:ijms22168403. [PMID: 34445108 PMCID: PMC8395079 DOI: 10.3390/ijms22168403] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
Atopic dermatitis (AD) is a common inflammatory dermatosis affecting up to 30% of children and 10% of adults worldwide. AD is primarily driven by an epidermal barrier defect which triggers immune dysregulation within the skin. According to recent research such phenomena are closely related to the microbial dysbiosis of the skin. There is growing evidence that cutaneous microbiota and bacterial biofilms negatively affect skin barrier function, contributing to the onset and exacerbation of AD. This review summarizes the latest data on the mechanisms leading to microbiome dysbiosis and biofilm formation in AD, and the influence of these phenomena on skin barrier function.
Collapse
Affiliation(s)
- Leszek Blicharz
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
- Correspondence:
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Joanna Czuwara
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Anna Waśkiel-Burnat
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Mohamad Goldust
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Małgorzata Olszewska
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Zbigniew Samochocki
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| |
Collapse
|
13
|
Yuan M, Wang X, Yan X, Ding H, Yi J, Xia H, Yu X. Effects of heat-sensitive moxibustion combined with naprapathy and warming needle moxibustion combined with naprapathy in patients with periarthritis of shoulder. Am J Transl Res 2021; 13:7804-7811. [PMID: 34377257 PMCID: PMC8340244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/03/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To evaluate the effects of heat-sensitive moxibustion (HSM) combined with naprapathy and warming needle moxibustion (WNM) combined with naprapathy on shoulder function and serum levels of calcitonin gene-related peptide (CGRP), substance P (SP), tumor necrosis factor-α (TNF-α) and interleukin-2 (IL-2) in patients with periarthritis of shoulder (POS). METHODS From July 2017 to July 2020, sixty patients with POS admitted to our hospital were selected as the study subjects, and divided into HSM group (n=29) receiving HSM combined with naprapathy and WNM group receiving WNM combined with naprapathy (n=31). The changes in shoulder function, degrees of pain and serum levels of CGRP, SP, TNF-α and IL-2 were compared between the two groups. RESULTS After treatment, the scores of myodynamia, pain, range of motion (ROM) of shoulder joint and activities of daily living (ADLs) were improved in both groups (P<0.05), and the scores in HSM group were remarkably higher than those in WNM group (P<0.05). Visual analogue scale (VAS) scores after 3 courses of treatment were lower than those after 1 and 2 courses of treatment respectively (P<0.05), and the VAS scores in HSM group were markedly lower than those in WNM group after 1, 2, and 3 courses of treatment (P<0.05). After treatment, the serum levels of CGRP, SP, TNF-α and IL-2 were decreased in both groups (P<0.05), and the levels in HSM group were noticeably lower than those in WNM group (P<0.05). CONCLUSION HSM combined with naprapathy is superior to WNM combined with naprapathy in inhibition of inflammatory factors of pain and serum inflammatory factors, alleviating the pain and promoting the restoration of shoulder function in patients with POS.
Collapse
Affiliation(s)
- Minghua Yuan
- Rehabilitation Department, Yichun People's Hospital Yichun 336000, Jiangxi, China
| | - Xiaoyuan Wang
- Rehabilitation Department, Yichun People's Hospital Yichun 336000, Jiangxi, China
| | - Xiaoyan Yan
- Rehabilitation Department, Yichun People's Hospital Yichun 336000, Jiangxi, China
| | - Hong Ding
- Rehabilitation Department, Yichun People's Hospital Yichun 336000, Jiangxi, China
| | - Jing Yi
- Rehabilitation Department, Yichun People's Hospital Yichun 336000, Jiangxi, China
| | - Hongmei Xia
- Rehabilitation Department, Yichun People's Hospital Yichun 336000, Jiangxi, China
| | - Xueqin Yu
- Rehabilitation Department, Yichun People's Hospital Yichun 336000, Jiangxi, China
| |
Collapse
|
14
|
Sweat metabolome and proteome: Recent trends in analytical advances and potential biological functions. J Proteomics 2021; 246:104310. [PMID: 34198014 DOI: 10.1016/j.jprot.2021.104310] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022]
Abstract
Metabolome and proteome profiling of biofluids, e.g., urine, plasma, has generated vast and ever-increasing amounts of knowledge over the last few decades. Paradoxically, omics analyses of sweat, one of the most readily available human biofluids, have lagged behind. This review capitalizes on the current knowledge and state of the art analytical advances of sweat metabolomics and proteomics. Moreover, current applications of sweat omics such as the discovery of disease biomarkers and monitoring athletic performance are also presented in this review. Another area of emerging knowledge that has been highlighted herein lies in the role of skin host-microbiome interactions in shaping the sweat metabolite-protein profiles. Discussion of future research directions describes the need to have a better grasp of sweat chemicals and to better understand how they function as aided by advances in omics tools. Overall, the role of sweat as an information-rich biofluid that could complement the exploration of the skin metabolome/proteome is emphasized.
Collapse
|
15
|
Mart’yanov SV, Botchkova EA, Plakunov VK, Gannesen AV. The Impact of Norepinephrine on Mono-Species and Dual-Species Staphylococcal Biofilms. Microorganisms 2021; 9:820. [PMID: 33924447 PMCID: PMC8070549 DOI: 10.3390/microorganisms9040820] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/01/2021] [Accepted: 04/11/2021] [Indexed: 01/17/2023] Open
Abstract
The effect of norepinephrine ("NE") on Gram-negative bacteria is well characterized; however, little is known about the impact of NE on cutaneous Gram-positive skin residents, especially staphylococci. In this study, the impact of NE on monospecies and dual-species biofilms of Staphylococcus epidermidis and S. aureus model strains was investigated for the first time. Biofilms were grown in two different models (on polytetrafluoroethylene ("PTFE") cubes and glass microfiber filters ("GMFFs")) and additionally kinetic measurements of bacterial growth was performed. We have shown that NE can affect the biofilm formation of both species with a strong dependence on aerobic or anaerobic culture conditions in different models. It was shown that S. epidermidis suppresses S. aureus growth in dual-species biofilms and that NE can accelerate this process, contributing to the competitive behavior of staphylococci.
Collapse
Affiliation(s)
- Sergey Vladislavovich Mart’yanov
- Laboratory of Viability of Microorganisms, Federal Research Center “Fundamentals of Biotechnology” of Russian Academy of Sciences, 117312 Moscow, Russia; (S.V.M.); (V.K.P.)
| | - Ekaterina Alexandrovna Botchkova
- Laboratory of Microbiology of Anthropogenic Habitats, Federal Research Center “Fundamentals of Biotechnology” of Russian Academy of Sciences, 117312 Moscow, Russia;
| | - Vladimir Konstantinovich Plakunov
- Laboratory of Viability of Microorganisms, Federal Research Center “Fundamentals of Biotechnology” of Russian Academy of Sciences, 117312 Moscow, Russia; (S.V.M.); (V.K.P.)
| | - Andrei Vladislavovich Gannesen
- Laboratory of Viability of Microorganisms, Federal Research Center “Fundamentals of Biotechnology” of Russian Academy of Sciences, 117312 Moscow, Russia; (S.V.M.); (V.K.P.)
| |
Collapse
|
16
|
Augustyniak D, Kramarska E, Mackiewicz P, Orczyk-Pawiłowicz M, Lundy FT. Mammalian Neuropeptides as Modulators of Microbial Infections: Their Dual Role in Defense versus Virulence and Pathogenesis. Int J Mol Sci 2021; 22:ijms22073658. [PMID: 33915818 PMCID: PMC8036953 DOI: 10.3390/ijms22073658] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
The regulation of infection and inflammation by a variety of host peptides may represent an evolutionary failsafe in terms of functional degeneracy and it emphasizes the significance of host defense in survival. Neuropeptides have been demonstrated to have similar antimicrobial activities to conventional antimicrobial peptides with broad-spectrum action against a variety of microorganisms. Neuropeptides display indirect anti-infective capacity via enhancement of the host’s innate and adaptive immune defense mechanisms. However, more recently concerns have been raised that some neuropeptides may have the potential to augment microbial virulence. In this review we discuss the dual role of neuropeptides, perceived as a double-edged sword, with antimicrobial activity against bacteria, fungi, and protozoa but also capable of enhancing virulence and pathogenicity. We review the different ways by which neuropeptides modulate crucial stages of microbial pathogenesis such as adhesion, biofilm formation, invasion, intracellular lifestyle, dissemination, etc., including their anti-infective properties but also detrimental effects. Finally, we provide an overview of the efficacy and therapeutic potential of neuropeptides in murine models of infectious diseases and outline the intrinsic host factors as well as factors related to pathogen adaptation that may influence efficacy.
Collapse
Affiliation(s)
- Daria Augustyniak
- Department of Pathogen Biology and Immunology, Faculty of Biology, University of Wroclaw, 51-148 Wroclaw, Poland;
- Correspondence: ; Tel.: +48-71-375-6296
| | - Eliza Kramarska
- Department of Pathogen Biology and Immunology, Faculty of Biology, University of Wroclaw, 51-148 Wroclaw, Poland;
- Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, 80134 Napoli, Italy
| | - Paweł Mackiewicz
- Department of Bioinformatics and Genomics, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland;
| | | | - Fionnuala T. Lundy
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK;
| |
Collapse
|
17
|
Fournière M, Latire T, Souak D, Feuilloley MGJ, Bedoux G. Staphylococcus epidermidis and Cutibacterium acnes: Two Major Sentinels of Skin Microbiota and the Influence of Cosmetics. Microorganisms 2020; 8:E1752. [PMID: 33171837 PMCID: PMC7695133 DOI: 10.3390/microorganisms8111752] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/26/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Dermatological and cosmetics fields have recently started to focus on the human skin microbiome and microbiota, since the skin microbiota is involved in the health and dysbiosis of the skin ecosystem. Amongst the skin microorganisms, Staphylococcus epidermidis and Cutibacterium acnes, both commensal bacteria, appear as skin microbiota sentinels. These sentinels have a key role in the skin ecosystem since they protect and prevent microbiota disequilibrium by fighting pathogens and participate in skin homeostasis through the production of beneficial bacterial metabolites. These bacteria adapt to changing skin microenvironments and can shift to being opportunistic pathogens, forming biofilms, and thus are involved in common skin dysbiosis, such as acne or atopic dermatitis. The current evaluation methods for cosmetic active ingredient development are discussed targeting these two sentinels with their assets and limits. After identification of these objectives, research of the active cosmetic ingredients and products that maintain and promote these commensal metabolisms, or reduce their pathogenic forms, are now the new challenges of the skincare industry in correlation with the constant development of adapted evaluation methods.
Collapse
Affiliation(s)
- Mathilde Fournière
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Bretagne Sud, 56000 Vannes, France; (T.L.); (G.B.)
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Catholique de l’Ouest Bretagne Nord, 22200 Guingamp, France
| | - Thomas Latire
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Bretagne Sud, 56000 Vannes, France; (T.L.); (G.B.)
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Catholique de l’Ouest Bretagne Nord, 22200 Guingamp, France
| | - Djouhar Souak
- Laboratoire de Microbiologie Signaux et Microenvironment LMSM EA4312, Université de Rouen Normandie, 27000 Évreux, France; (D.S.); (M.G.J.F.)
- BASF Beauty Care Solutions France SAS, 69007 Lyon, France
| | - Marc G. J. Feuilloley
- Laboratoire de Microbiologie Signaux et Microenvironment LMSM EA4312, Université de Rouen Normandie, 27000 Évreux, France; (D.S.); (M.G.J.F.)
| | - Gilles Bedoux
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Bretagne Sud, 56000 Vannes, France; (T.L.); (G.B.)
| |
Collapse
|
18
|
The Role of the Microbiome and Microbiome-Derived Metabolites in Atopic Dermatitis and Non-Histaminergic Itch. Am J Clin Dermatol 2020; 21:44-50. [PMID: 32910440 PMCID: PMC7584541 DOI: 10.1007/s40257-020-00538-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent advances in our understanding of the pathophysiology of atopic dermatitis (AD) have revealed that skin microbiome dysbiosis plays an important role in the disease. In this review, we describe how changes in the structure and function of the microbiome are involved in the pathogenesis of AD. We highlight recent data showing that differential changes in microbial diversity, both within and across communities from different body habitats (including the skin, gut, and oral mucosa), are associated with the development and severity of AD. We also describe recent evidence demonstrating that the metabolic activity of the skin microbiome can act as a regulator of inflammation, with alterations in the level of a skin microbiome-derived tryptophan metabolite, indole-3-aldehyde (IAId), being shown to play a role in AD. The various mechanisms by which interactions between the microbiome and components of the non-histaminergic pathway result in itch in AD are also discussed.
Collapse
|
19
|
Danilova ND, Solovyeva TV, Mart’yanov SV, Zhurina MV, Gannesen AV. Stimulatory Effect of Epinephrine on Biofilms of Micrococcus luteus C01. Microbiology (Reading) 2020. [DOI: 10.1134/s0026261720040049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
20
|
Sung Kim H, Yosipovitch G. THE SKIN MICROBIOTA AND ITCH: Is There a Link? THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2020; 13:S39-S46. [PMID: 33282109 PMCID: PMC7710288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Itch is an unpleasant sensation that emanates primarily from the skin. The chemical mediators that drive neuronal activity originate from a complex interaction between keratinocytes, inflammatory cells, nerve endings, and the skin microbiota, relaying itch signals to the brain. Stress also exacerbates itch via the skin-brain axis. Recently, the microbiota has surfaced as a major player to regulate this axis, notably during stress settings aroused by actual or perceived homeostatic challenge. The routes of communication between the microbiota and brain are slowly being unraveled and involve neurochemicals (i.e., acetylcholine, histamine, catecholamines, and corticotropin) that originate from the microbiota itself. By focusing on itch biology and by referring to the more established field of pain research, this review examines the possible means by which the skin microbiota contributes to itch.
Collapse
Affiliation(s)
- Hei Sung Kim
- Dr. Kim is with the Department of Dermatology and Cutaneous Surgery at Miami Itch Center, Miller School of Medicine at University of Miami in Miami, Florida, the Department of Dermatology at Incheon St. Mary's Hospital, The Catholic University of Korea in Seoul, Korea, and the Department of Biomedicine and Health Sciences, at The Catholic University of Korea in Seoul, Korea
- Dr. Yosipovitch is with the Department of Dermatology and Cutaneous Surgery at Miami Itch Center, Miller School of Medicine at the University of Miami in Miami, Florida
| | - Gil Yosipovitch
- Dr. Kim is with the Department of Dermatology and Cutaneous Surgery at Miami Itch Center, Miller School of Medicine at University of Miami in Miami, Florida, the Department of Dermatology at Incheon St. Mary's Hospital, The Catholic University of Korea in Seoul, Korea, and the Department of Biomedicine and Health Sciences, at The Catholic University of Korea in Seoul, Korea
- Dr. Yosipovitch is with the Department of Dermatology and Cutaneous Surgery at Miami Itch Center, Miller School of Medicine at the University of Miami in Miami, Florida
| |
Collapse
|
21
|
The Skin Microbiota and Itch: Is There a Link? J Clin Med 2020; 9:jcm9041190. [PMID: 32331207 PMCID: PMC7230651 DOI: 10.3390/jcm9041190] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Itch is an unpleasant sensation that emanates primarily from the skin. The chemical mediators that drive neuronal activity originate from a complex interaction between keratinocytes, inflammatory cells, nerve endings and the skin microbiota, relaying itch signals to the brain. Stress also exacerbates itch via the skin–brain axis. Recently, the microbiota has surfaced as a major player to regulate this axis, notably during stress settings aroused by actual or perceived homeostatic challenge. The routes of communication between the microbiota and brain are slowly being unraveled and involve neurochemicals (i.e., acetylcholine, histamine, catecholamines, corticotropin) that originate from the microbiota itself. By focusing on itch biology and by referring to the more established field of pain research, this review examines the possible means by which the skin microbiota contributes to itch.
Collapse
|
22
|
T cell receptor cross-reactivity between gliadin and bacterial peptides in celiac disease. Nat Struct Mol Biol 2019; 27:49-61. [PMID: 31873306 DOI: 10.1038/s41594-019-0353-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/18/2019] [Indexed: 12/16/2022]
Abstract
The human leukocyte antigen (HLA) locus is strongly associated with T cell-mediated autoimmune disorders. HLA-DQ2.5-mediated celiac disease (CeD) is triggered by the ingestion of gluten, although the relative roles of genetic and environmental risk factors in CeD is unclear. Here we identify microbially derived mimics of gliadin epitopes and a parental bacterial protein that is naturally processed by antigen-presenting cells and activated gliadin reactive HLA-DQ2.5-restricted T cells derived from CeD patients. Crystal structures of T cell receptors in complex with HLA-DQ2.5 bound to two distinct bacterial peptides demonstrate that molecular mimicry underpins cross-reactivity toward the gliadin epitopes. Accordingly, gliadin reactive T cells involved in CeD pathogenesis cross-react with ubiquitous bacterial peptides, thereby suggesting microbial exposure as a potential environmental factor in CeD.
Collapse
|
23
|
Characterization of Reactive and Sensitive Skin Microbiota: Effect of Halymenia durvillei (HD) Extract Treatment. COSMETICS 2019. [DOI: 10.3390/cosmetics6040069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
After characterization of the reactive skin microbiota, we investigated whether the active Halymenia durvillei (HD), rich in polysaccharides, could modulate this microbiota after 28 days of treatment, act on neuroinflammation parameters, and calm feelings of discomfort and redness. Skin microbiota was assessed using next-generation sequencing experiments (16S RNA gene fragment sequencing) on samples collected from 30 volunteers suffering from reactive, sensitive skin. To evaluate the effect of the HD extract on neuroinflammation, we used an ex vivo model. Finally, an in vivo study was performed using a clinical assessment (blood microcirculation via videocapillaroscopy) of functional signs employing the Sensitive Scale and the soothing effect was evaluated and compared to a placebo treatment. At the phylum level, the samples were mostly composed of Actinobacteria, Proteobacteria, Firmicutes, and Bacteroidetes, which accounted for more than 97% of the total sequencing read in all samples, with no differences before or after treatment with the HD active ingredient. The Shannon Diversity index indicated lower microbial communities compared to healthy skin. Maintenance of the Shannon Diversity index was reported after 28 days of HD active ingredient treatment, wherein microbial communities continued to decrease in number during treatment with the placebo. The average taxonomic composition of associated skin microbial communities showed that reactive skin is characterized by a low proportion of the Chryseobacterium genus compared to a high proportion of the Corynebacterium genus. At the species level, Actinobacteria are mainly represented by Propionibacterium acnes (72.13%) and Corynebacterium kroppenstedtii (13.23%), representing species typically observed in clinical cases of redness, the main criteria for volunteer inclusion. Corynebacterium kroppenstedtii, with increased levels being associated with skin redness, decreased with HD treatment. This decrease coincided with the clinical improvement observed after 7 weeks of treatment. The ex vivo study revealed that the HD extract induced a significant decrease in the expression of TRPV-1 (−67%; p < 0.001) and NK1-R (−43%; p < 0.01) compared to the control after 6 days of treatment. These data support the use of polysaccharides, found in red alga, in the treatment of reactive and sensitive skin related to the modulation of skin microbiota.
Collapse
|
24
|
Borrel V, Thomas P, Catovic C, Racine PJ, Konto-Ghiorghi Y, Lefeuvre L, Duclairoir-Poc C, Zouboulis CC, Feuilloley MGJ. Acne and Stress: Impact of Catecholamines on Cutibacterium acnes. Front Med (Lausanne) 2019; 6:155. [PMID: 31355200 PMCID: PMC6635461 DOI: 10.3389/fmed.2019.00155] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 06/21/2019] [Indexed: 12/22/2022] Open
Abstract
Cutibacterium acnes (former Propionibacterium acnes), is a bacterium characterized by high genomic variability, consisting of four subtypes and six major ribotypes. Skin is the largest neuroendocrine organ of the human body and many cutaneous hormones and neurohormones can modulate bacterial physiology. Here, we investigated the effect of catecholamines, i.e., epinephrine and norepinephrine, on two representative strains of C. acnes, of which the genome has been fully sequenced, identified as RT4 acneic and RT6 non-acneic strains. Epinephrine and norepinephrine (10-6 M) had no impact on the growth of C. acnes but epinephrine increased RT4 and RT6 biofilm formation, as measured by crystal violet staining, whereas norepinephrine was only active on the RT4 strain. We obtained the same results by confocal microscopy with the RT4 strain, whereas there was no effect of either catecholamine on the RT6 strain. However, this strain was also sensitive to catecholamines, as shown by MATs tests, as epinephrine and norepinephrine affected its surface polarity. Flow cytometry studies revealed that epinephrine and norepinephrine are unable to induce major changes of bacterial surface properties and membrane integrity. Exposure of sebocytes to control or catecholamine-treated bacteria showed epinephrine and norepinephrine to have no effect on the cytotoxic or inflammatory potential of either C. acnes strains but to stimulate their effect on sebocyte lipid synthesis. Uriage thermal spring water was previously shown to inhibit biofilm production by C. acnes. We thus tested its effect after exposure of the bacteria to epinephrine and norepinephrine. The effect of the thermal water on the response of C. acnes to catecholamines depended on the surface on which the biofilm was grown. Finally, an in-silico study revealed the presence of a protein in the genome of C. acnes that shows homology with the catecholamine receptor of Escherichia coli and eukaryotes. This study suggests that C. acnes may play a role as a relay between stress mediators (catecholamines) and acne.
Collapse
Affiliation(s)
- Valérie Borrel
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Pauline Thomas
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Chloé Catovic
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Pierre-Jean Racine
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Yoan Konto-Ghiorghi
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Luc Lefeuvre
- R&D Uriage Dermatological Laboratory, Neuilly sur Seine, France
| | - Cécile Duclairoir-Poc
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Christos C Zouboulis
- Departments of Dermatology, Venereology, Allergology, and Immunology, Dessau Medical Center, Brandenburg Medical School Theodor Fontane, Dessau, Germany
| | - Marc G J Feuilloley
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| |
Collapse
|
25
|
Mechanism of action of the moonlighting protein EfTu as a Substance P sensor in Bacillus cereus. Sci Rep 2019; 9:1304. [PMID: 30718605 PMCID: PMC6361937 DOI: 10.1038/s41598-018-37506-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/30/2018] [Indexed: 01/20/2023] Open
Abstract
The striking feature of the ubiquitous protein EfTu (Thermo unstable ribosomal Elongation factor) is its moonlighting (multifunctional) activity. Beyond its function at the ribosomal level it should be exported to the bacterial surface and act as an environmental sensor. In Bacillus cereus, and other cutaneous bacteria, it serves as a Substance P (SP) receptor and is essential for bacterial adaptation to the host. However, the modus operandi of EfTu as a bacterial sensor remains to be investigated. Studies realized by confocal and transmission electron microscopy revealed that, in the absence of an exogenous signal, EfTu is not exposed on the bacterial surface but is recruited under the effect of SP. In addition, SP acts as a transcriptional regulator of the tuf gene encoding for EfTu. As observed using gadolinium chloride, an inhibitor of membrane mechanosensitive channels (Msc), Msc control EfTu export and subsequently the bacterial response to SP both in terms of cytotoxicity and biofilm formation activity. Microscale thermophoresis revealed that in response to SP, EfTu can form homopolymers. This event should occur after EfTu export and, as shown by proteo-liposome reconstruction studies, SP appears to promote EfTu polymers association to the membrane, leading subsequently to the bacterial response. Molecular modeling suggests that this mechanism should involve EfTu unfolding and insertion into the bacterial cytoplasmic membrane, presumably through formation of homopolymers. This study is unraveling the original mechanism action of EfTu as a bacterial sensor but also reveals that this protein should have a broader role, including in eukaryotes.
Collapse
|
26
|
Lesouhaitier O, Clamens T, Rosay T, Desriac F, Louis M, Rodrigues S, Gannesen A, Plakunov VK, Bouffartigues E, Tahrioui A, Bazire A, Dufour A, Cornelis P, Chevalier S, Feuilloley MGJ. Host Peptidic Hormones Affecting Bacterial Biofilm Formation and Virulence. J Innate Immun 2018; 11:227-241. [PMID: 30396172 PMCID: PMC6738206 DOI: 10.1159/000493926] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 12/25/2022] Open
Abstract
Bacterial biofilms constitute a critical problem in hospitals, especially in resuscitation units or for immunocompromised patients, since bacteria embedded in their own matrix are not only protected against antibiotics but also develop resistant variant strains. In the last decade, an original approach to prevent biofilm formation has consisted of studying the antibacterial potential of host communication molecules. Thus, some of these compounds have been identified for their ability to modify the biofilm formation of both Gram-negative and Gram-positive bacteria. In addition to their effect on biofilm production, a detailed study of the mechanism of action of these human hormones on bacterial physiology has allowed the identification of new bacterial pathways involved in biofilm formation. In this review, we focus on the impact of neuropeptidic hormones on bacteria, address some future therapeutic issues, and provide a new view of inter-kingdom communication.
Collapse
Affiliation(s)
- Olivier Lesouhaitier
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France,
| | - Thomas Clamens
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France
| | - Thibaut Rosay
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France
| | - Florie Desriac
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France
| | - Mélissande Louis
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France
| | - Sophie Rodrigues
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France
| | - Andrei Gannesen
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France
- Winogradsky Institute of Microbiology, Research Center of Biotechnology of RAS, Moscow, Russian Federation
- Lomonosov Moscow State University, Moscow, Russian Federation
| | - Vladimir K Plakunov
- Winogradsky Institute of Microbiology, Research Center of Biotechnology of RAS, Moscow, Russian Federation
| | - Emeline Bouffartigues
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France
| | - Ali Tahrioui
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France
| | - Alexis Bazire
- Laboratoire de Biotechnologie et Chimie Marines EA 3884, IUEM, Université de Bretagne-Sud (UBL), Lorient, France
| | - Alain Dufour
- Laboratoire de Biotechnologie et Chimie Marines EA 3884, IUEM, Université de Bretagne-Sud (UBL), Lorient, France
| | - Pierre Cornelis
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France
| | - Sylvie Chevalier
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France
| | - Marc G J Feuilloley
- Laboratory of Microbiology Signals and Microenvironment, LMSM EA 4312, Normandy University, University of Rouen Normandy, Evreux, France
| |
Collapse
|
27
|
Antidromic neurogenic activity and cutaneous bacterial flora. Semin Immunopathol 2018; 40:281-289. [DOI: 10.1007/s00281-018-0671-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 03/06/2018] [Indexed: 12/13/2022]
|
28
|
Desvaux M, Candela T, Serror P. Surfaceome and Proteosurfaceome in Parietal Monoderm Bacteria: Focus on Protein Cell-Surface Display. Front Microbiol 2018; 9:100. [PMID: 29491848 PMCID: PMC5817068 DOI: 10.3389/fmicb.2018.00100] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 01/16/2018] [Indexed: 12/12/2022] Open
Abstract
The cell envelope of parietal monoderm bacteria (archetypal Gram-positive bacteria) is formed of a cytoplasmic membrane (CM) and a cell wall (CW). While the CM is composed of phospholipids, the CW is composed at least of peptidoglycan (PG) covalently linked to other biopolymers, such as teichoic acids, polysaccharides, and/or polyglutamate. Considering the CW is a porous structure with low selective permeability contrary to the CM, the bacterial cell surface hugs the molecular figure of the CW components as a well of the external side of the CM. While the surfaceome corresponds to the totality of the molecules found at the bacterial cell surface, the proteinaceous complement of the surfaceome is the proteosurfaceome. Once translocated across the CM, secreted proteins can either be released in the extracellular milieu or exposed at the cell surface by associating to the CM or the CW. Following the gene ontology (GO) for cellular components, cell-surface proteins at the CM can either be integral (GO: 0031226), i.e., the integral membrane proteins, or anchored to the membrane (GO: 0046658), i.e., the lipoproteins. At the CW (GO: 0009275), cell-surface proteins can be covalently bound, i.e., the LPXTG-proteins, or bound through weak interactions to the PG or wall polysaccharides, i.e., the cell wall binding proteins. Besides monopolypeptides, some proteins can associate to each other to form supramolecular protein structures of high molecular weight, namely the S-layer, pili, flagella, and cellulosomes. After reviewing the cell envelope components and the different molecular mechanisms involved in protein attachment to the cell envelope, perspectives in investigating the proteosurfaceome in parietal monoderm bacteria are further discussed.
Collapse
Affiliation(s)
- Mickaël Desvaux
- Université Clermont Auvergne, INRA, UMR454 MEDiS, Clermont-Ferrand, France
| | - Thomas Candela
- EA4043 Unité Bactéries Pathogènes et Santé, Châtenay-Malabry, France
| | - Pascale Serror
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
29
|
Song JX, Ren H, Gao YF, Lee CY, Li SF, Zhang F, Li L, Chen H. Dietary Capsaicin Improves Glucose Homeostasis and Alters the Gut Microbiota in Obese Diabetic ob/ob Mice. Front Physiol 2017; 8:602. [PMID: 28890700 PMCID: PMC5575157 DOI: 10.3389/fphys.2017.00602] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 08/04/2017] [Indexed: 12/13/2022] Open
Abstract
Background: The effects of capsaicin on obesity and glucose homeostasis are still controversial and the mechanisms underlying these effects remain largely unknown. This study aimed to investigate the potential relationship between the regulation of obesity and glucose homeostasis by dietary capsaicin and the alterations of gut microbiota in obese diabetic ob/ob mice. Methods: The ob/ob mice were subjected to a normal, low-capsaicin (0.01%), or high-capsaicin (0.02%) diet for 6 weeks, respectively. Obesity phenotypes, glucose homeostasis, the gut microbiota structure and composition, short-chain fatty acids, gastrointestinal hormones, and pro-inflammatory cytokines were measured. Results: Both the low- and high-capsaicin diets failed to prevent the increase in body weight, adiposity index, and Lee's obesity index. However, dietary capsaicin at both the low and high doses significantly inhibited the increase of fasting blood glucose and insulin levels. These inhibitory effects were comparable between the two groups. Similarly, dietary capsaicin resulted in remarkable improvement in glucose and insulin tolerance. In addition, neither the low- nor high-capsaicin diet could alter the α-diversity and β-diversity of the gut microbiota. Taxonomy-based analysis showed that both the low- and high-capsaicin diets, acting in similar ways, significantly increased the Firmicutes/Bacteroidetes ratio at the phylum level as well as increased the Roseburia abundance and decreased the Bacteroides and Parabacteroides abundances at the genus level. Spearman's correlation analysis revealed that the Roseburia abundance was negatively while the Bacteroides and Parabacteroides abundances were positively correlated to the fasting blood glucose level and area under the curve by the oral glucose tolerance test. Finally, the low- and high-capsaicin diets significantly increased the fecal butyrate and plasma total GLP-1 levels, but decreased plasma total ghrelin, TNF-α, IL-1β, and IL-6 levels as compared with the normal diet. Conclusions: The beneficial effects of dietary capsaicin on glucose homeostasis are likely associated with the alterations of specific bacteria at the genus level. These alterations in bacteria induced by dietary capsaicin contribute to improved glucose homeostasis through increasing short-chain fatty acids, regulating gastrointestinal hormones and inhibiting pro-inflammatory cytokines. However, our results should be interpreted cautiously due to the lower caloric intake at the initial stage after capsaicin diet administration.
Collapse
Affiliation(s)
- Jun-Xian Song
- Department of Cardiology, Peking University People's HospitalBeijing, China.,Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's HospitalBeijing, China.,Center for Cardiovascular Translational Research, Peking University People's HospitalBeijing, China
| | - Hui Ren
- Department of Cardiology, Peking University People's HospitalBeijing, China.,Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's HospitalBeijing, China.,Center for Cardiovascular Translational Research, Peking University People's HospitalBeijing, China
| | - Yuan-Feng Gao
- Department of Cardiology, Peking University People's HospitalBeijing, China.,Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's HospitalBeijing, China.,Center for Cardiovascular Translational Research, Peking University People's HospitalBeijing, China
| | - Chong-You Lee
- Department of Cardiology, Peking University People's HospitalBeijing, China.,Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's HospitalBeijing, China.,Center for Cardiovascular Translational Research, Peking University People's HospitalBeijing, China
| | - Su-Fang Li
- Department of Cardiology, Peking University People's HospitalBeijing, China.,Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's HospitalBeijing, China.,Center for Cardiovascular Translational Research, Peking University People's HospitalBeijing, China
| | - Feng Zhang
- Department of Cardiology, Peking University People's HospitalBeijing, China.,Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's HospitalBeijing, China.,Center for Cardiovascular Translational Research, Peking University People's HospitalBeijing, China
| | - Long Li
- Department of Cardiology, Peking University People's HospitalBeijing, China.,Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's HospitalBeijing, China.,Center for Cardiovascular Translational Research, Peking University People's HospitalBeijing, China
| | - Hong Chen
- Department of Cardiology, Peking University People's HospitalBeijing, China.,Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's HospitalBeijing, China.,Center for Cardiovascular Translational Research, Peking University People's HospitalBeijing, China
| |
Collapse
|