1
|
Whitmore M, Tobin I, Burkardt A, Zhang G. Nutritional Modulation of Host Defense Peptide Synthesis: A Novel Host-Directed Antimicrobial Therapeutic Strategy? Adv Nutr 2024; 15:100277. [PMID: 39053604 PMCID: PMC11381887 DOI: 10.1016/j.advnut.2024.100277] [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: 02/28/2024] [Revised: 06/11/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
The escalating threat of antimicrobial resistance underscores the imperative for innovative therapeutic strategies. Host defense peptides (HDPs), integral components of innate immunity, exhibit profound antimicrobial and immunomodulatory properties. Various dietary compounds, such as short-chain fatty acids, vitamins, minerals, sugars, amino acids, phytochemicals, bile acids, probiotics, and prebiotics have been identified to enhance the synthesis of endogenous HDPs without provoking inflammatory response or compromising barrier integrity. Additionally, different classes of these compounds synergize in augmenting HDP synthesis and disease resistance. Moreover, dietary supplementation of several HDP-inducing compounds or their combinations have demonstrated robust protection in rodents, rabbits, pigs, cattle, and chickens from experimental infections. However, the efficacy of these compounds in inducing HDP synthesis varies considerably among distinct compounds. Additionally, the regulation of HDP genes occurs in a gene-specific, cell type-specific, and species-specific manner. In this comprehensive review, we systematically summarized the modulation of HDP synthesis and the mechanism of action attributed to each major class of dietary compounds, including their synergistic combinations, across a spectrum of animal species including humans. We argue that the ability to enhance innate immunity and barrier function without triggering inflammation or microbial resistance positions the nutritional modulation of endogenous HDP synthesis as a promising host-directed approach for mitigating infectious diseases and antimicrobial resistance. These HDP-inducing compounds, particularly in combinations, harbor substantial clinical potential for further exploration in antimicrobial therapies for both human and other animals.
Collapse
Affiliation(s)
- Melanie Whitmore
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Isabel Tobin
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Amanda Burkardt
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States.
| |
Collapse
|
2
|
Morio KA, Sternowski RH, Brogden KA. Induction of Endogenous Antimicrobial Peptides to Prevent or Treat Oral Infection and Inflammation. Antibiotics (Basel) 2023; 12:antibiotics12020361. [PMID: 36830272 PMCID: PMC9952314 DOI: 10.3390/antibiotics12020361] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
Antibiotics are often used to treat oral infections. Unfortunately, excessive antibiotic use can adversely alter oral microbiomes and promote the development of antibiotic-resistant microorganisms, which can be difficult to treat. An alternate approach could be to induce the local transcription and expression of endogenous oral antimicrobial peptides (AMPs). To assess the feasibility and benefits of this approach, we conducted literature searches to identify (i) the AMPs expressed in the oral cavity; (ii) the methods used to induce endogenous AMP expression; and (iii) the roles that expressed AMPs may have in regulating oral inflammation, immunity, healing, and pain. Search results identified human neutrophil peptides (HNP), human beta defensins (HBD), and cathelicidin AMP (CAMP) gene product LL-37 as prominent AMPs expressed by oral cells and tissues. HNP, HBD, and LL-37 expression can be induced by micronutrients (trace elements, elements, and vitamins), nutrients, macronutrients (mono-, di-, and polysaccharides, amino acids, pyropeptides, proteins, and fatty acids), proinflammatory agonists, thyroid hormones, and exposure to ultraviolet (UV) irradiation, red light, or near infrared radiation (NIR). Localized AMP expression can help reduce infection, inflammation, and pain and help oral tissues heal. The use of a specific inducer depends upon the overall objective. Inducing the expression of AMPs through beneficial foods would be suitable for long-term health protection. Additionally, the specialized metabolites or concentrated extracts that are utilized as dosage forms would maintain the oral and intestinal microbiome composition and control oral and intestinal infections. Inducing AMP expression using irradiation methodologies would be applicable to a specific oral treatment area in addition to controlling local infections while regulating inflammatory and healing processes.
Collapse
Affiliation(s)
| | | | - Kim A. Brogden
- College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
- Correspondence:
| |
Collapse
|
3
|
Shannon AH, Adelman SA, Hisey EA, Potnis SS, Rozo V, Yung MW, Li JY, Murphy CJ, Thomasy SM, Leonard BC. Antimicrobial Peptide Expression at the Ocular Surface and Their Therapeutic Use in the Treatment of Microbial Keratitis. Front Microbiol 2022; 13:857735. [PMID: 35722307 PMCID: PMC9201425 DOI: 10.3389/fmicb.2022.857735] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/12/2022] [Indexed: 11/29/2022] Open
Abstract
Microbial keratitis is a common cause of ocular pain and visual impairment worldwide. The ocular surface has a relatively paucicellular microbial community, mostly found in the conjunctiva, while the cornea would be considered relatively sterile. However, in patients with microbial keratitis, the cornea can be infected with multiple pathogens including Staphylococcus aureus, Pseudomonas aeruginosa, and Fusarium sp. Treatment with topical antimicrobials serves as the standard of care for microbial keratitis, however, due to high rates of pathogen resistance to current antimicrobial medications, alternative therapeutic strategies must be developed. Multiple studies have characterized the expression and activity of antimicrobial peptides (AMPs), endogenous peptides with key antimicrobial and wound healing properties, on the ocular surface. Recent studies and clinical trials provide promise for the use of AMPs as therapeutic agents. This article reviews the repertoire of AMPs expressed at the ocular surface, how expression of these AMPs can be modulated, and the potential for harnessing the AMPs as potential therapeutics for patients with microbial keratitis.
Collapse
Affiliation(s)
- Allison H. Shannon
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Sara A. Adelman
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Erin A. Hisey
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Sanskruti S. Potnis
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Vanessa Rozo
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Madeline W. Yung
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Jennifer Y. Li
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Christopher J. Murphy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Sara M. Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Brian C. Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
4
|
Chang Y, Mei J, Yang T, Zhang Z, Liu G, Zhao H, Chen X, Tian G, Cai J, Wu B, Wu F, Jia G. Effect of Dietary Zinc Methionine Supplementation on Growth Performance, Immune Function and Intestinal Health of Cherry Valley Ducks Challenged With Avian Pathogenic Escherichia coli. Front Microbiol 2022; 13:849067. [PMID: 35602082 PMCID: PMC9115567 DOI: 10.3389/fmicb.2022.849067] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
This study was carried out to evaluate the effects of supplemental zinc methionine (Zn-Met) on growth performance, immune function, and intestinal health of meat ducks challenged with avian pathogenic Escherichia coli (APEC). A total of 480 1-day-old Cherry Valley male ducks were randomly assigned to 8 treatments with 10 replicates, each replicate containing 10 ducks. A 4 × 2 factor design was used with four dietary zinc levels (0, 30, 60, 120 mg Zn/kg in the form Zn-Met was added to the corn-soybean basal diet) and challenged with or without APEC at 8-days-old ducks. The trial lasted for 14 days. The results showed that a dietary Zn-Met supplementation significantly increased body weight (BW) of 14 days and BW gain, and decreased mortality during 7-14-days-old ducks (p < 0.05). Furthermore, dietary 30, 60, 120 mg/kg Zn-Met supplementation noticeably increased the thymus index at 2 days post-infection (2 DPI) and 8 DPI (p < 0.05), and 120 mg/kg Zn-Met enhanced the serum IgA at 2 DPI and IgA, IgG, IgM, C3 at 8 DPI (p < 0.05). In addition, dietary 120 mg/kg Zn-Met supplementation dramatically increased villus height and villus height/crypt depth (V/C) of jejunum at 2 DPI and 8 DPI (p < 0.05). The TNF-α and IFN-γ mRNA expression were downregulated after supplemented with 120 mg/kg Zn-Met in jejunum at 8 DPI (p < 0.05). Moreover, dietary 120 mg/kg Zn-Met supplementation stimulated ZO-3, OCLN mRNA expression at 2 DPI and ZO-2 mRNA expression in jejunum at 8 DPI (p < 0.05), and improved the MUC2 concentration in jejunum at 2 DPI and 8 DPI (p < 0.05). At the same time, the cecal Bifidobacterium and Lactobacillus counts were increased (p < 0.05), and Escherichia coli counts were decreased (p < 0.05) after supplemented with Zn-Met. In conclusion, inclusion of 120 mg/kg Zn-Met minimizes the adverse effects of APEC challenge on meat ducks by improving growth performance and enhancing immune function and intestinal health.
Collapse
Affiliation(s)
- Yaqi Chang
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Jia Mei
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Ting Yang
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang, China
| | - Zhenyu Zhang
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Institute of Animal Husbandry and Veterinary Medicine, Meishan Vocational Technical College, Meishan, China
| | - Guangmang Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Hua Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Gang Tian
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Jingyi Cai
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Bing Wu
- Chelota Group, Guanghan, China
| | - Fali Wu
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Gang Jia
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
5
|
Oh HJ, Park YJ, Cho JH, Song MH, Gu BH, Yun W, Lee JH, An JS, Kim YJ, Lee JS, Kim S, Kim H, Kim ES, Lee BK, Kim BW, Kim HB, Cho JH, Kim MH. Changes in Diarrhea Score, Nutrient Digestibility, Zinc Utilization, Intestinal Immune Profiles, and Fecal Microbiome in Weaned Piglets by Different Forms of Zinc. Animals (Basel) 2021; 11:ani11051356. [PMID: 34064626 PMCID: PMC8151337 DOI: 10.3390/ani11051356] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Piglets, especially at the weaning stage, are highly susceptible to various diseases due to an incomplete immune system development and stress responses. Post-weaning diarrhea has a significant impact on piglet growth rate and mortality, resulting in economic losses to the swine industry. Zinc oxide (ZnO) is widely used as a weaning diet supplement in the swine industry to prevent diarrheal diseases and promote immune system development. Despite the recently demonstrated beneficial effects of ZnO, many efforts have been made to reduce its excessive use in piglets owing to environmental pollution and toxic effects on tissues; thus, the need for an effective alternative ZnO form, which promotes zinc utilization, has been gaining attention. However, we do not completely understand the mode of action of ZnO alternatives or the amount required to exert positive effects on weaned piglets. Therefore, we conducted this study to evaluate the effects of different forms of ZnO alternatives (ZnO chelate with glycine (chelate-ZnO) and nanoparticle-sized ZnO (nano-ZnO)) on diarrhea score, nutrient digestibility, zinc utilization, intestinal immune profiles, and fecal microflora on piglets, together with a comparison of the standard ZnO treatment. We found that 200 ppm Nano-ZnO had similar positive effects on weaned piglets compared with 2500 ppm ZnO and therefore is a promising alternative to ZnO. Abstract Twenty weaned piglets with initial body weight of 6.83 ± 0.33 kg (21 day of age, LYD) were randomly assigned to four treatments for a two-week feeding trial to determine the effects of different dietary zinc on nutrient digestibility, intestinal health, and microbiome of weaned piglets. The dietary treatments included a negative control (CON), standard ZnO (ZnO, 2500 ppm), zinc chelate with glycine (Chelate-ZnO, 200 ppm), and nanoparticle-sized ZnO (Nano-ZnO, 200 ppm). At 0 to 1 week, the diarrhea score was decreased in the CON group compared with the ZnO, Chelate-ZnO, and Nano-ZnO group. In overall period, the ZnO and Nano-ZnO groups exhibited improved diarrhea scores compared to the CON group. The apparent total tract digestibility of dry matter and gross energy was the lowest in the CON group after one week. Compared to the ZnO group, the chelate-ZnO group exhibited higher proportion of T-bet+ and FoxP3+ T cells and the nano-ZnO group had higher numbers of RORgt+ and GATA3+ T cells in the mesenteric lymph nodes. ZnO group increased IL-6 and IL-8 levels in the colon tissues and these positive effects were observed in both chelate ZnO and nano-ZnO groups with lower level. The 16S rRNA gene analysis showed that the relative abundance of Prevotella was higher in the ZnO-treated groups than in the CON group and that of Succinivibrio was the highest in the nano-ZnO group. The relative abundance of Lactobacillus increased in the ZnO group. In conclusion, low nano-ZnO levels have similar effects on nutrient digestibility, fecal microflora, and intestinal immune profiles in weaning pigs; thus, nano-ZnO could be used as a ZnO alternative for promoting ZnO utilization and intestinal immunity.
Collapse
Affiliation(s)
- Han-Jin Oh
- Department of Animal Sciences, Chungbuk National University, Cheongju 286-44, Korea; (H.-J.O.); (W.Y.); (J.-H.L.); (J.-S.A.); (Y.-J.K.); (J.-S.L.); (B.-K.L.)
| | - Yei-Ju Park
- Department of Animal Sciences, Pusan National University, Miryang 50463, Korea; (Y.-J.P.); (B.-W.K.)
| | - Jae Hyoung Cho
- Department of Animal Resource, and Science, Dankook University, Cheonan 311-16, Korea; (J.H.C.); (S.K.); (H.K.); (E.S.K.)
| | - Min-Ho Song
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 341-34, Korea;
| | - Bon-Hee Gu
- Life and Industry Convergence Research Institute, Pusan National University, Mirayng 50463, Korea;
| | - Won Yun
- Department of Animal Sciences, Chungbuk National University, Cheongju 286-44, Korea; (H.-J.O.); (W.Y.); (J.-H.L.); (J.-S.A.); (Y.-J.K.); (J.-S.L.); (B.-K.L.)
| | - Ji-Hwan Lee
- Department of Animal Sciences, Chungbuk National University, Cheongju 286-44, Korea; (H.-J.O.); (W.Y.); (J.-H.L.); (J.-S.A.); (Y.-J.K.); (J.-S.L.); (B.-K.L.)
| | - Ji-Seon An
- Department of Animal Sciences, Chungbuk National University, Cheongju 286-44, Korea; (H.-J.O.); (W.Y.); (J.-H.L.); (J.-S.A.); (Y.-J.K.); (J.-S.L.); (B.-K.L.)
| | - Yong-Ju Kim
- Department of Animal Sciences, Chungbuk National University, Cheongju 286-44, Korea; (H.-J.O.); (W.Y.); (J.-H.L.); (J.-S.A.); (Y.-J.K.); (J.-S.L.); (B.-K.L.)
| | - Jun-Soeng Lee
- Department of Animal Sciences, Chungbuk National University, Cheongju 286-44, Korea; (H.-J.O.); (W.Y.); (J.-H.L.); (J.-S.A.); (Y.-J.K.); (J.-S.L.); (B.-K.L.)
| | - Sheena Kim
- Department of Animal Resource, and Science, Dankook University, Cheonan 311-16, Korea; (J.H.C.); (S.K.); (H.K.); (E.S.K.)
| | - Hyeri Kim
- Department of Animal Resource, and Science, Dankook University, Cheonan 311-16, Korea; (J.H.C.); (S.K.); (H.K.); (E.S.K.)
| | - Eun Sol Kim
- Department of Animal Resource, and Science, Dankook University, Cheonan 311-16, Korea; (J.H.C.); (S.K.); (H.K.); (E.S.K.)
| | - Byoung-Kon Lee
- Department of Animal Sciences, Chungbuk National University, Cheongju 286-44, Korea; (H.-J.O.); (W.Y.); (J.-H.L.); (J.-S.A.); (Y.-J.K.); (J.-S.L.); (B.-K.L.)
| | - Byeong-Woo Kim
- Department of Animal Sciences, Pusan National University, Miryang 50463, Korea; (Y.-J.P.); (B.-W.K.)
- Life and Industry Convergence Research Institute, Pusan National University, Mirayng 50463, Korea;
| | - Hyeun Bum Kim
- Department of Animal Resource, and Science, Dankook University, Cheonan 311-16, Korea; (J.H.C.); (S.K.); (H.K.); (E.S.K.)
- Correspondence: (H.B.K.); (J.-H.C.); (M.-H.K.); Tel.: +82-043-261-2544 (H.B.K.); +82-041-550-3652 (J.-H.C.); +82-55-350-5516 (M.-H.K.); Fax: +82-043-273-2240 (H.B.K.); +82-041-550-3604 (J.-H.C.); +82-55-350-5519 (M.-H.K.)
| | - Jin-Ho Cho
- Department of Animal Sciences, Chungbuk National University, Cheongju 286-44, Korea; (H.-J.O.); (W.Y.); (J.-H.L.); (J.-S.A.); (Y.-J.K.); (J.-S.L.); (B.-K.L.)
- Correspondence: (H.B.K.); (J.-H.C.); (M.-H.K.); Tel.: +82-043-261-2544 (H.B.K.); +82-041-550-3652 (J.-H.C.); +82-55-350-5516 (M.-H.K.); Fax: +82-043-273-2240 (H.B.K.); +82-041-550-3604 (J.-H.C.); +82-55-350-5519 (M.-H.K.)
| | - Myung-Hoo Kim
- Department of Animal Sciences, Pusan National University, Miryang 50463, Korea; (Y.-J.P.); (B.-W.K.)
- Life and Industry Convergence Research Institute, Pusan National University, Mirayng 50463, Korea;
- Correspondence: (H.B.K.); (J.-H.C.); (M.-H.K.); Tel.: +82-043-261-2544 (H.B.K.); +82-041-550-3652 (J.-H.C.); +82-55-350-5516 (M.-H.K.); Fax: +82-043-273-2240 (H.B.K.); +82-041-550-3604 (J.-H.C.); +82-55-350-5519 (M.-H.K.)
| |
Collapse
|
6
|
Wang Y, Wang M, Shan A, Feng X. Avian host defense cathelicidins: structure, expression, biological functions, and potential therapeutic applications. Poult Sci 2020; 99:6434-6445. [PMID: 33248558 PMCID: PMC7704953 DOI: 10.1016/j.psj.2020.09.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 08/14/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022] Open
Abstract
Host defense peptides (HDP) are multifunctional effectors of the innate immune system, which has antimicrobial and pleiotropic immunomodulatory functions. Although there is a very sophisticated superposition of adaptive immune systems in vertebrates, this system is still essential. As an important family of HDP, cathelicidins are also known for their broad-spectrum antibacterial activity against bacteria, fungi, and enveloped viruses. It has been found in humans and other species, including cattle, pigs, sheep, goats, chickens, rabbits, and some kind of fish. Among them, cathelicidins in birds were described for the first time in 2005. This review focuses on the structure, biological activities, expression, and regulation of avian cathelicidin, especially main effects of host defense cathelicidin on potential therapeutic applications. According to the results obtained both in vitro and in vivo, good perspectives have been opened for cathelicidin. Nevertheless, further studies are needed to better characterize the mechanisms of action underlying the beneficial effects of cathelicidin as novel therapeutic alternatives to antibiotics.
Collapse
Affiliation(s)
- Yingjie Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Min Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Xingjun Feng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, People's Republic of China.
| |
Collapse
|
7
|
Abstract
Background Enthusiasm for the use of metal nanoparticles in human and veterinary medicine is high. Many articles describe the effects of metal nanoparticles on microbes in vitro, and a smaller number of articles describe effects on the immune system, which is the focus of this review. Methods Articles were retrieved by performing literature searches in Medline, of the National Institute of Medicine, as well as via Google Scholar. Results In vitro studies show that metal nanoparticles have antimicrobial effects. Some metal nanoparticles augment innate host immune defenses, such as endogenous antimicrobial peptides, and nitric oxide. Metal nanoparticles may also function as vaccine adjuvants. Metal nanoparticles can migrate to locations distant from the site of administration, however, requiring careful monitoring for toxicity. Conclusions Metal nanoparticles show a great deal of potential as immunomodulators, as well as direct antimicrobial effects. Before metal particles can be adopted as therapies; however, more studies are needed to determine how nanoparticles migrate though the body and on possible adverse effects.
Collapse
Affiliation(s)
- John K Crane
- Division of Infectious Diseases, University at Buffalo , Buffalo, New York, USA
| |
Collapse
|
8
|
Wu J, Ma N, Johnston LJ, Ma X. Dietary Nutrients Mediate Intestinal Host Defense Peptide Expression. Adv Nutr 2020; 11:92-102. [PMID: 31204774 PMCID: PMC7442325 DOI: 10.1093/advances/nmz057] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/14/2019] [Accepted: 05/17/2019] [Indexed: 02/06/2023] Open
Abstract
The intestinal tract is the shared locus of intestinal epithelial cells, immune cells, nutrient digestion and absorption, and microbial survival. The gut in animals faces continuous challenges in communicating with the external environment. Threats from endogenous imbalance and exogenous feeds, especially pathogens, could trigger a disorder of homeostasis, leading to intestinal disease and even systematic disease risk. As a part of the intestinal protective barrier, endogenous host defense peptides (HDPs) play multiple beneficial physiological roles in the gut mucosa. Moreover, enhancing endogenous HDPs is being developed as a new strategy for resisting pathogens and commensal microbes, and to maintain intestinal health and reduce antibiotic use. In recent years, multiple nutrients such as branched-chain amino acids, SCFAs, lactose, zinc, and cholecalciferol (vitamin D3) have been reported to significantly increase HDP expression. Nutritional intervention has received more attention and is viewed as a promising means to defend against pathogenic infections and intestinal inflammation. The present review focuses on current discoveries surrounding HDP expression and nutritional regulation of mechanisms in the gut. Our aim is to provide a comprehensive overview, referable tactics, and novel opinions.
Collapse
Affiliation(s)
- Jianmin Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ning Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lee J Johnston
- Swine Nutrition and Production, West Central Research and Outreach Center, University of Minnesota, Morris, MN, USA
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Department of Internal Medicine, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
9
|
Chen J, Zhai Z, Long H, Yang G, Deng B, Deng J. Inducible expression of defensins and cathelicidins by nutrients and associated regulatory mechanisms. Peptides 2020; 123:170177. [PMID: 31704211 DOI: 10.1016/j.peptides.2019.170177] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
Abstract
Host defense peptides (HDPs) are crucial components of the body's first line of defense that protect organisms from infections and mediate immune responses. Defensins and cathelicidins are the two most important families of HDPs in mammals. In this review, we summarize the nutrients that are involved in inducible expression of endogenous defensins and cathelicidins. In addition, the mitogen-activated protein kinases (MAPK), nuclear factor kappa B (NF-κB) and histone deacetylase (HDAC) signaling pathways that play vital roles in the induction of defensin and cathelicidin expression are highlighted. Endogenous defensins and cathelicidins induced by nutrients may be potential alternatives to antibiotic treatments against infection and diseases. This review mainly focuses on the inducible expression and regulatory mechanisms of defensins and cathelicidins in multiple species by different nutrients and the potential applications of defensin- and cathelicidin-inducing nutrients.
Collapse
Affiliation(s)
- Jialuo Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhenya Zhai
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Hongrong Long
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Guangming Yang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Baichuan Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
10
|
Lemaire J, Mireault M, Jumarie C. Zinc interference with Cd‐induced hormetic effect in differentiated Caco‐2 cells: Evidence for inhibition downstream ERK activation. J Biochem Mol Toxicol 2019; 34:e22437. [DOI: 10.1002/jbt.22437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/07/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Joannie Lemaire
- Département des Sciences Biologiques, groupe TOXENUniversité du Québec à MontréalMontréal Québec Canada
| | - Myriam Mireault
- Département des Sciences Biologiques, groupe TOXENUniversité du Québec à MontréalMontréal Québec Canada
| | - Catherine Jumarie
- Département des Sciences Biologiques, groupe TOXENUniversité du Québec à MontréalMontréal Québec Canada
| |
Collapse
|
11
|
Wang J, Zhang W, Wang S, Liu H, Zhang D, Wang Y, Ji H. Swine-Derived Probiotic Lactobacillus plantarum Modulates Porcine Intestinal Endogenous Host Defense Peptide Synthesis Through TLR2/MAPK/AP-1 Signaling Pathway. Front Immunol 2019; 10:2691. [PMID: 31803195 PMCID: PMC6877743 DOI: 10.3389/fimmu.2019.02691] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/31/2019] [Indexed: 01/29/2023] Open
Abstract
Host defense peptides (HDPs) have antimicrobial and immunoregulatory activities and are involved in epithelial innate immune defense. Dietary modulation of endogenous HDP synthesis is an effective way to boost the host innate immune system. This study aimed to investigate the role of the probiotic Lactobacillus plantarum strain ZLP001 in porcine HDP induction and the underlying mechanism. To this end, we evaluated the stimulatory effect of L. plantarum ZLP001 on HDP expression in piglet intestinal tissue in vivo and porcine IPEC-J2 cells and 3D4/31 cells in vitro, and we examined the underlying intracellular signaling pathway in IPEC-J2 cells. L. plantarum ZLP001 treatment increased the mRNA expression of jejunal and ileal HDPs in weaned piglets. In IPEC-J2 and 3D4/31 cells, L. plantarum ZLP001 stimulated HDP expression, but different HDP induction patterns were observed, with the various HDPs exhibiting different relative mRNA levels in each cell line. L. plantarum ZLP001 induced porcine HDP expression through toll-like receptor (TLR)2 recognition as evidenced by the fact that HDP expression was suppressed in TLR2-knockdown IPEC-J2 cells. Furthermore, we found that L. plantarum ZLP001 activated the extracellular signal-regulated kinase (ERK)1/2 and c-jun N-terminal kinase (JNK) signaling pathways, as indicated by enhanced phosphorylation of both ERK1/2 and JNK and the fact that HDP expression was suppressed upon inhibition of ERK1/2 and JNK. Furthermore, L. plantarum ZLP001 activated c-fos and c-jun transcription factor phosphorylation and activity. We conclude that L. plantarum ZLP001 induces porcine HDP expression in vivo and in vitro, and the induction seems to be regulated via TLR2 as well as the ERK1/2/JNK and c-jun/c-fos signaling pathways. Modulation of endogenous HDPs mediated by L. plantarum ZLP001 might be a promising approach to improving intestinal health and enhancing diarrhea resistance in weaning piglets.
Collapse
Affiliation(s)
- Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Wei Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Sixin Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Hui Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Dongyan Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Yamin Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Haifeng Ji
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| |
Collapse
|
12
|
Wang J, Ji H, Wang S, Liu H, Zhang W, Zhang D, Wang Y. Probiotic Lactobacillus plantarum Promotes Intestinal Barrier Function by Strengthening the Epithelium and Modulating Gut Microbiota. Front Microbiol 2018; 9:1953. [PMID: 30197632 PMCID: PMC6117384 DOI: 10.3389/fmicb.2018.01953] [Citation(s) in RCA: 258] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/02/2018] [Indexed: 12/15/2022] Open
Abstract
Weaning disturbs the intestinal barrier function and increases the risk of infection in piglets. Probiotics exert beneficial health effects, mainly by reinforcing the intestinal epithelium and modulating the gut microbiota. However, the mechanisms of action, and especially, the specific regulatory effects of modulated microbiota by probiotics on the intestinal epithelium have not yet been elucidated. The present study aimed to decipher the protective effects of the probiotic Lactobacillus plantarum strain ZLP001 on the intestinal epithelium and microbiota as well as the effects of modulated microbiota on epithelial function. Paracellular permeability was measured with fluorescein isothiocyanate-dextran (FD-4). Gene and protein expression levels of tight junction (TJ) proteins, proinflammatory cytokines, and host defense peptides were determined by RT-qPCR, ELISA, and western blot analysis. Short-chain fatty acid (SCFA) concentrations were measured by ion chromatography. Fecal microbiota composition was assessed by high-throughput sequencing. The results showed that pretreatment with 108 colony forming units (CFU) mL−1 of L. plantarum ZLP001 significantly counteracted the increase in gut permeability to FD-4 induced by 106 CFU mL−1 enterotoxigenic Escherichia coli (ETEC). In addition, L. plantarum ZLP001 pretreatment alleviated the reduction in TJ proteins (claudin-1, occludin, and ZO-1) and downregulated proinflammatory cytokines IL-6 and IL-8, and TNFα expression and secretion caused by ETEC. L. plantarum ZLP001 also significantly increased the expression of the host defense peptides pBD2 and PG1-5 and pBD2 secretion relative to the control. Furthermore, L. plantarum ZLP001 treatment affected piglet fecal microbiota. The abundance of butyrate-producing bacteria Anaerotruncus and Faecalibacterium was significantly increased in L. plantarum ZLP001-treated piglets, and showed a positive correlation with fecal butyric and acetic acid concentrations. In addition, the cell density of Clostridium sensu stricto 1, which may cause epithelial inflammation, was decreased after L. plantarum ZLP001 administration, while the beneficial Lactobacillus was significantly increased. Our findings suggest that L. plantarum ZLP001 fortifies the intestinal barrier by strengthening epithelial defense functions and modulating gut microbiota.
Collapse
Affiliation(s)
- Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Haifeng Ji
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Sixin Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Hui Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Wei Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Dongyan Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Yamin Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| |
Collapse
|
13
|
Wang J, Zeng Y, Wang S, Liu H, Zhang D, Zhang W, Wang Y, Ji H. Swine-Derived Probiotic Lactobacillus plantarum Inhibits Growth and Adhesion of Enterotoxigenic Escherichia coli and Mediates Host Defense. Front Microbiol 2018; 9:1364. [PMID: 29997590 PMCID: PMC6028558 DOI: 10.3389/fmicb.2018.01364] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/05/2018] [Indexed: 01/28/2023] Open
Abstract
Weaning stress renders piglets susceptible to pathogen infection, which leads to post-weaning diarrhea, a severe condition characterized by heavy diarrhea and mortality in piglets. Enterotoxigenic Escherichia coli (ETEC) is one of typical strains associated with post-weaning diarrhea. Thus, prevention and inhibition of ETEC infection are of great concern. Probiotics possess anti-pathogenic activity and can counteract ETEC infection; however, their underlying mechanisms and modes of action have not yet been clarified. In the present study, the direct and indirect protective effects of Lactobacillus plantarum ZLP001 against ETEC infection were investigated by different methods. We found that bacterial culture and culture supernatant of L. plantarum ZLP001 prevented ETEC growth by the Oxford cup method, and ETEC growth inhibition was observed in a co-culture assay as well. This effect was suggested to be caused mainly by antimicrobial metabolites produced by L. plantarum ZLP001. In addition, adhesion capacity of L. plantarum ZLP001 to IPEC-J2 cells were observed using microscopy and counting. L. plantarum ZLP001 also exhibited a concentration-dependent ability to inhibit ETEC adhesion to IPEC-J2 cells, which mainly occurred via exclusion and competition mode. Furthermore, quantitative real time polymerase chain reaction (qPCR) analysis showed that L. plantarum ZLP001 upregulated the expression of host defense peptides (HDPs) but did not trigger an inflammatory response. In addition, L. plantarum ZLP001 induced HDP secretion, which enhanced the potential antimicrobial activity of IPEC-J2 cell-culture supernatant after incubation with L. plantarum ZLP001. Our findings demonstrate that L. plantarum ZLP001, an intestinal Lactobacillus species associated with piglet health, possesses anti-ETEC activity. L. plantarum ZLP001 might prevent ETEC growth, inhibit ETEC adhesion to the intestinal mucosa, and activate the innate immune response to secret antimicrobial peptides. L. plantarum ZLP001 is worth investigation as a potential probiotics.
Collapse
Affiliation(s)
- Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Yanxia Zeng
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Sixin Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Hui Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Dongyan Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Wei Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Yamin Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Haifeng Ji
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| |
Collapse
|
14
|
Rahman MT, Karim MM. Metallothionein: a Potential Link in the Regulation of Zinc in Nutritional Immunity. Biol Trace Elem Res 2018; 182:1-13. [PMID: 28585004 DOI: 10.1007/s12011-017-1061-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/22/2017] [Indexed: 01/03/2023]
Abstract
Nutritional immunity describes mechanisms for withholding essential transition metals as well as directing the toxicity of these metals against infectious agents. Zinc is one of these transition elements that are essential for both humans and microbial pathogens. At the same time, Zn can be toxic both for man and microbes if its concentration is higher than the tolerance limit. Therefore a "delicate" balance of Zn must be maintained to keep the immune cells surveilling while making the level of Zn either to starve or to intoxicate the pathogens. On the other hand, the invading pathogens will exploit the host Zn pool for its survival and replication. Apparently, different sets of protein in human and bacteria are involved to maintain their Zn need. Metallothionein (MT)-a group of low molecular weight proteins, is well known for its Zn-binding ability and is expected to play an important role in that Zn balance at the time of active infection. However, the differences in structural, functional, and molecular control of biosynthesis between human and bacterial MT might play an important role to determine the proper use of Zn and the winning side. The current review explains the possible involvement of human and bacterial MT at the time of infection to control and exploit Zn for their need.
Collapse
|
15
|
Mechanistic insights into the protective impact of zinc on sepsis. Cytokine Growth Factor Rev 2017; 39:92-101. [PMID: 29279185 DOI: 10.1016/j.cytogfr.2017.12.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/19/2017] [Indexed: 12/11/2022]
Abstract
Sepsis, a systemic inflammation as a response to a bacterial infection, is a huge unmet medical need. Data accumulated over the last decade suggest that the nutritional status of patients as well as composition of their gut microbiome, are strongly linked with the risk to develop sepsis, the severity of the disease and prognosis. In particular, the essential micronutrient zinc is essential in the resistance against sepsis and has shown to be protective in animal models as well as in human patients. The potential mechanisms by which zinc protects in sepsis are discussed in this review paper: we will focus on the inflammatory response, chemotaxis, phagocytosis, immune response, oxidative stress and modulation of the microbiome. A full understanding of the mechanism of action of zinc may open new preventive and therapeutic interventions in sepsis.
Collapse
|
16
|
Liu H, Hou C, Wang G, Jia H, Yu H, Zeng X, Thacker PA, Zhang G, Qiao S. Lactobacillus reuteri I5007 Modulates Intestinal Host Defense Peptide Expression in the Model of IPEC-J2 Cells and Neonatal Piglets. Nutrients 2017; 9:nu9060559. [PMID: 28561758 PMCID: PMC5490538 DOI: 10.3390/nu9060559] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 12/15/2022] Open
Abstract
Modulation of the synthesis of endogenous host defense peptides (HDPs) by probiotics represents a novel antimicrobial approach for disease control and prevention, particularly against antibiotic-resistant infections in human and animals. However, the extent of HDP modulation by probiotics is species dependent and strain specific. In the present study, The porcine small intestinal epithelial cell line (IPEC-J2) cells and neonatal piglets were used as in-vitro and in-vivo models to test whether Lactobacillus reuteri I5007 could modulate intestinal HDP expression. Gene expressions of HDPs, toll-like receptors, and fatty acid receptors were determined, as well as colonic short chain fatty acid concentrations and microbiota. Exposure to 108 colony forming units (CFU)/mL of L. reuteri I5007 for 6 h significantly increased the expression of porcine β-Defensin2 (PBD2), pBD3, pBD114, pBD129, and protegrins (PG) 1-5 in IPEC-J2 cells. Similarly, L. reuteri I5007 administration significantly increased the expression of jejunal pBD2 as well as colonic pBD2, pBD3, pBD114, and pBD129 in neonatal piglets (p < 0.05). This was probably associated with the increase in colonic butyric acid concentration and up-regulating expression of Peroxisome Proliferator-Activated Receptor-γ (PPAR-γ) and G Protein-Coupled Receptor 41 (GPR41) (p < 0.05), but not with stimulation of Pattern-Recognition Receptors. Additionally, supplementation with L. reuteri I5007 in the piglets did not affect the colonic microbiota structure. Our findings suggested that L. reuteri I5007 could modulate intestinal HDP expression and improve the gut health of neonatal piglets, probably through the increase in colonic butyric acid concentration and the up-regulation of the downstream molecules of butyric acid, PPAR-γ and GPR41, but not through modifying gut microbiota structure.
Collapse
Affiliation(s)
- Hongbin Liu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.
| | - Chengli Hou
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.
- Institute of Food Science and Technology CAAS, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Gang Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.
| | - Hongmin Jia
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.
| | - Haitao Yu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.
| | - Philip A Thacker
- Department of Animal and Poultry Science, University of Saskatchewan, SK S7N 5C5, Canada.
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74074, USA.
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
17
|
Alcayaga-Miranda F, Cuenca J, Khoury M. Antimicrobial Activity of Mesenchymal Stem Cells: Current Status and New Perspectives of Antimicrobial Peptide-Based Therapies. Front Immunol 2017; 8:339. [PMID: 28424688 PMCID: PMC5371613 DOI: 10.3389/fimmu.2017.00339] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/09/2017] [Indexed: 12/20/2022] Open
Abstract
While mesenchymal stem cells (MSCs)-based therapy appears to be promising, there are concerns regarding possible side effects related to the unwanted suppression of antimicrobial immunity leading to an increased risk of infection. Conversely, recent data show that MSCs exert strong antimicrobial effects through indirect and direct mechanisms, partially mediated by the secretion of antimicrobial peptides and proteins (AMPs). In fact, MSCs have been reported to increase bacterial clearance in preclinical models of sepsis, acute respiratory distress syndrome, and cystic fibrosis-related infections. This article reviews the current evidence regarding the direct antimicrobial effector function of MSCs, focusing mainly on the role of MSCs-derived AMPs. The strategies that might modulate the expression and secretion of these AMPs, leading to enhanced antimicrobial effect, are highlighted. Furthermore, studies evaluating the presence of AMPs in the cargo of extracellular vesicles (EVs) are underlined as perspective opportunities to develop new drug delivery tools. The antimicrobial potential of MSCs-derived EVs can also be heightened through cell conditioning and/or drug loading. Finally, improving the pharmacokinetics and delivery, in addition to deciphering the multi-target drug status of AMPs, should synergistically lead to key advances against infections caused by drug-resistant strains.
Collapse
Affiliation(s)
- Francisca Alcayaga-Miranda
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile
| | - Jimena Cuenca
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile
| | - Maroun Khoury
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile.,Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| |
Collapse
|
18
|
Mullin JM, Diguilio KM, Valenzano MC, Deis R, Thomas S, Zurbach EP, Abdulhaqq S, Montaner LJ. Zinc reduces epithelial barrier compromise induced by human seminal plasma. PLoS One 2017; 12:e0170306. [PMID: 28278250 PMCID: PMC5344308 DOI: 10.1371/journal.pone.0170306] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 01/03/2017] [Indexed: 01/11/2023] Open
Abstract
Human semen has the potential to modulate the epithelial mucosal tissues it contacts, as seminal plasma (SP) is recognized to contain both pro- and anti-barrier components, yet its effects on epithelial barrier function are largely unknown. We addressed the role of human SP when exposed to the basal-lateral epithelial surface, a situation that would occur clinically with prior mechanical or disease-related injury of the human epithelial mucosal cell layers in contact with semen. The action of SP on claudins-2, -4, -5, and -7 expression, as well as on a target epithelium whose basolateral surface has been made accessible to SP, showed upregulation of claudins-4 and -5 in CACO-2 human epithelial cell layers, despite broad variance in SP-induced modulation of transepithelial electrical resistance and mannitol permeability. Upregulation of claudin-2 by SP also exhibited such variance by SP sample. We characterize individual effects on CACO-2 barrier function of nine factors known to be present abundantly in seminal plasma (zinc, EGF, citrate, spermine, fructose, urea, TGF, histone, inflammatory cytokines) to establish that zinc, spermine and fructose had significant potential to raise CACO-2 transepithelial resistance, whereas inflammatory cytokines and EGF decreased this measure of barrier function. The role of zinc as a dominant factor in determining higher levels of transepithelial resistance and lower levels of paracellular leak were confirmed by zinc chelation and exogenous zinc addition. As expected, SP presentation to the basolateral cell surface also caused a very dramatic yet transient elevation of pErk levels. Results suggest that increased zinc content in SP can compete against the barrier-compromising effect of negative modulators in SP when SP gains access to that epithelium's basolateral surface. Prophylactic elevation of zinc in an epithelial cell layer prior to contact by SP may help to protect an epithelial barrier from invasion by SP-containing STD microbial pathogens such as HPV or HIV.
Collapse
Affiliation(s)
- James M. Mullin
- Lankenau Institute for Medical Research, Wynnewood, PA, United States of America
- * E-mail:
| | | | - Mary C. Valenzano
- Lankenau Institute for Medical Research, Wynnewood, PA, United States of America
| | - Rachael Deis
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | - Sunil Thomas
- Lankenau Institute for Medical Research, Wynnewood, PA, United States of America
| | - E. Peter Zurbach
- Department of Chemistry, Saint Joseph’s University, Philadelphia, PA, United States of America
| | | | - Luis J. Montaner
- The Wistar Institute, Philadelphia, PA, United States of America
| |
Collapse
|
19
|
Jarosz M, Olbert M, Wyszogrodzka G, Młyniec K, Librowski T. Antioxidant and anti-inflammatory effects of zinc. Zinc-dependent NF-κB signaling. Inflammopharmacology 2017; 25:11-24. [PMID: 28083748 PMCID: PMC5306179 DOI: 10.1007/s10787-017-0309-4] [Citation(s) in RCA: 393] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 12/31/2016] [Indexed: 12/13/2022]
Abstract
Zinc is a nutritionally fundamental trace element, essential to the structure and function of numerous macromolecules, including enzymes regulating cellular processes and cellular signaling pathways. The mineral modulates immune response and exhibits antioxidant and anti-inflammatory activity. Zinc retards oxidative processes on a long-term basis by inducing the expression of metallothioneins. These metal-binding cysteine-rich proteins are responsible for maintaining zinc-related cell homeostasis and act as potent electrophilic scavengers and cytoprotective agents. Furthermore, zinc increases the activation of antioxidant proteins and enzymes, such as glutathione and catalase. On the other hand, zinc exerts its antioxidant effect via two acute mechanisms, one of which is the stabilization of protein sulfhydryls against oxidation. The second mechanism consists in antagonizing transition metal-catalyzed reactions. Zinc can exchange redox active metals, such as copper and iron, in certain binding sites and attenuate cellular site-specific oxidative injury. Studies have demonstrated that physiological reconstitution of zinc restrains immune activation, whereas zinc deficiency, in the setting of severe infection, provokes a systemic increase in NF-κB activation. In vitro studies have shown that zinc decreases NF-κB activation and its target genes, such as TNF-α and IL-1β, and increases the gene expression of A20 and PPAR-α, the two zinc finger proteins with anti-inflammatory properties. Alternative NF-κB inhibitory mechanism is initiated by the inhibition of cyclic nucleotide phosphodiesterase, whereas another presumed mechanism consists in inhibition of IκB kinase in response to infection by zinc ions that have been imported into cells by ZIP8.
Collapse
Affiliation(s)
- Magdalena Jarosz
- Department of Radioligands, Jagiellonian University Medical College, Medyczna 9, 30-688, Krakow, Poland.
| | - Magdalena Olbert
- Department of Radioligands, Jagiellonian University Medical College, Medyczna 9, 30-688, Krakow, Poland
| | - Gabriela Wyszogrodzka
- Department of Pharmaceutical Technology and Biopharmaceutics, Jagiellonian University Medical College, Medyczna 9, 30-688, Krakow, Poland
| | - Katarzyna Młyniec
- Department of Pharmacobiology, Jagiellonian University Medical College, Medyczna 9, 30-688, Krakow, Poland
| | - Tadeusz Librowski
- Department of Radioligands, Jagiellonian University Medical College, Medyczna 9, 30-688, Krakow, Poland
| |
Collapse
|
20
|
Alcayaga-Miranda F, Cuenca J, Khoury M. Antimicrobial Activity of Mesenchymal Stem Cells: Current Status and New Perspectives of Antimicrobial Peptide-Based Therapies. Front Immunol 2017. [PMID: 28424688 DOI: 10.3389/fimmu.2017.0033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
While mesenchymal stem cells (MSCs)-based therapy appears to be promising, there are concerns regarding possible side effects related to the unwanted suppression of antimicrobial immunity leading to an increased risk of infection. Conversely, recent data show that MSCs exert strong antimicrobial effects through indirect and direct mechanisms, partially mediated by the secretion of antimicrobial peptides and proteins (AMPs). In fact, MSCs have been reported to increase bacterial clearance in preclinical models of sepsis, acute respiratory distress syndrome, and cystic fibrosis-related infections. This article reviews the current evidence regarding the direct antimicrobial effector function of MSCs, focusing mainly on the role of MSCs-derived AMPs. The strategies that might modulate the expression and secretion of these AMPs, leading to enhanced antimicrobial effect, are highlighted. Furthermore, studies evaluating the presence of AMPs in the cargo of extracellular vesicles (EVs) are underlined as perspective opportunities to develop new drug delivery tools. The antimicrobial potential of MSCs-derived EVs can also be heightened through cell conditioning and/or drug loading. Finally, improving the pharmacokinetics and delivery, in addition to deciphering the multi-target drug status of AMPs, should synergistically lead to key advances against infections caused by drug-resistant strains.
Collapse
Affiliation(s)
- Francisca Alcayaga-Miranda
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
- Cells for Cells, Santiago, Chile
| | - Jimena Cuenca
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
- Cells for Cells, Santiago, Chile
| | - Maroun Khoury
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
- Cells for Cells, Santiago, Chile
- Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| |
Collapse
|
21
|
Chromek M. The role of the antimicrobial peptide cathelicidin in renal diseases. Pediatr Nephrol 2015; 30:1225-32. [PMID: 25159719 DOI: 10.1007/s00467-014-2895-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 06/17/2014] [Accepted: 06/23/2014] [Indexed: 12/18/2022]
Abstract
The balance between the human body and surrounding microorganisms is crucial for homeostasis and health. A disturbance in host-pathogen interactions causes disease. Two important diseases of the kidney and urinary tract are directly caused by bacteria or bacterial toxins: urinary tract infection (UTI) and diarrhea-associated hemolytic uremic syndrome (HUS). In the majority of cases, UTIs are caused by bacteria ascending from the perineum through the urethra to the urinary tract. In contrast, HUS is caused by non-invasive bacteria, such as enterohemorrhagic Escherichia coli, which colonize the gut and do not enter the blood stream. In this latter case, the bacteria release Shiga toxin, which binds to blood cells and thus reaches the target organs, mainly kidneys. Interactions between Shiga toxin, blood cells and endothelial cells in the kidney lead to cell apoptosis and inflammation. Innate immunity and the antimicrobial peptide cathelicidin seem to play important roles in the pathogenesis of both UTI and HUS. Moreover, influencing cathelicidin production and release might offer new therapeutic and prophylactic strategies for both diseases.
Collapse
Affiliation(s)
- Milan Chromek
- Paediatric Nephrology Unit, Astrid Lindgren Children's Hospital, Karolinska University Hospital Huddinge, B57, SE-14186, Stockholm, Sweden,
| |
Collapse
|
22
|
Kuroda K, Okumura K, Isogai H, Isogai E. The Human Cathelicidin Antimicrobial Peptide LL-37 and Mimics are Potential Anticancer Drugs. Front Oncol 2015; 5:144. [PMID: 26175965 PMCID: PMC4485164 DOI: 10.3389/fonc.2015.00144] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 06/15/2015] [Indexed: 01/08/2023] Open
Abstract
Antimicrobial peptides (AMPs) play a critical role in innate host defense against microbial pathogens in many organisms. The human cathelicidin, LL-37, has a net positive charge and is amphiphilic, and can eliminate pathogenic microbes directly via electrostatic attraction toward negatively charged bacterial membranes. A number of studies have shown that LL-37 participates in various host immune systems, such as inflammatory responses and tissue repair, in addition to its antibacterial properties. Moreover, recent evidence suggests that it is also involved in the regulation of cancer. Indeed, previous studies have suggested that human LL-37 is involved in carcinogenesis via multiple reporters, such as FPR2 (FPRL1), epidermal growth factor receptor, and ERBb2, although LL-37 and its fragments and analogs also show anticancer effects in various cancer cell lines. This discrepancy can be attributed to peptide-based factors, host membrane-based factors, and signal regulation. Here, we describe the association between AMPs and cancer with a focus on anticancer peptide functions and selectivity in an effort to understand potential therapeutic implications.
Collapse
Affiliation(s)
- Kengo Kuroda
- Laboratory of Animal Microbiology, Graduate School of Agricultural Science, Tohoku University , Sendai , Japan
| | - Kazuhiko Okumura
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Health Sciences University of Hokkaido , Hokkaido , Japan
| | - Hiroshi Isogai
- Animal Research Center, Sapporo Medical University , Sapporo , Japan
| | - Emiko Isogai
- Laboratory of Animal Microbiology, Graduate School of Agricultural Science, Tohoku University , Sendai , Japan
| |
Collapse
|
23
|
Wang G. Human antimicrobial peptides and proteins. Pharmaceuticals (Basel) 2014; 7:545-94. [PMID: 24828484 PMCID: PMC4035769 DOI: 10.3390/ph7050545] [Citation(s) in RCA: 362] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 04/15/2014] [Accepted: 04/29/2014] [Indexed: 12/11/2022] Open
Abstract
As the key components of innate immunity, human host defense antimicrobial peptides and proteins (AMPs) play a critical role in warding off invading microbial pathogens. In addition, AMPs can possess other biological functions such as apoptosis, wound healing, and immune modulation. This article provides an overview on the identification, activity, 3D structure, and mechanism of action of human AMPs selected from the antimicrobial peptide database. Over 100 such peptides have been identified from a variety of tissues and epithelial surfaces, including skin, eyes, ears, mouths, gut, immune, nervous and urinary systems. These peptides vary from 10 to 150 amino acids with a net charge between -3 and +20 and a hydrophobic content below 60%. The sequence diversity enables human AMPs to adopt various 3D structures and to attack pathogens by different mechanisms. While α-defensin HD-6 can self-assemble on the bacterial surface into nanonets to entangle bacteria, both HNP-1 and β-defensin hBD-3 are able to block cell wall biosynthesis by binding to lipid II. Lysozyme is well-characterized to cleave bacterial cell wall polysaccharides but can also kill bacteria by a non-catalytic mechanism. The two hydrophobic domains in the long amphipathic α-helix of human cathelicidin LL-37 lays the basis for binding and disrupting the curved anionic bacterial membrane surfaces by forming pores or via the carpet model. Furthermore, dermcidin may serve as ion channel by forming a long helix-bundle structure. In addition, the C-type lectin RegIIIα can initially recognize bacterial peptidoglycans followed by pore formation in the membrane. Finally, histatin 5 and GAPDH(2-32) can enter microbial cells to exert their effects. It appears that granulysin enters cells and kills intracellular pathogens with the aid of pore-forming perforin. This arsenal of human defense proteins not only keeps us healthy but also inspires the development of a new generation of personalized medicine to combat drug-resistant superbugs, fungi, viruses, parasites, or cancer. Alternatively, multiple factors (e.g., albumin, arginine, butyrate, calcium, cyclic AMP, isoleucine, short-chain fatty acids, UV B light, vitamin D, and zinc) are able to induce the expression of antimicrobial peptides, opening new avenues to the development of anti-infectious drugs.
Collapse
Affiliation(s)
- Guangshun Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA.
| |
Collapse
|
24
|
Jiang W, Sunkara LT, Zeng X, Deng Z, Myers SM, Zhang G. Differential regulation of human cathelicidin LL-37 by free fatty acids and their analogs. Peptides 2013; 50:129-38. [PMID: 24140860 DOI: 10.1016/j.peptides.2013.10.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 10/08/2013] [Accepted: 10/09/2013] [Indexed: 11/19/2022]
Abstract
LL-37 is the single cathelicidin host defense peptide in humans with direct antimicrobial and immunomodulatory activities. Specific regulation of LL-37 synthesis has emerged as a novel non-antibiotic approach to disease control and prevention. Short-chain fatty acids, and butyrate in particular, were found recently to be strong inducers of LL-37 gene expression without causing inflammation. Here, we further evaluated the LL-37-inducing efficiency of a broad range of saturated free fatty acids and their derivatives in human HT-29 colonic epithelial cells and U-937 monocytic cells by real-time RT-PCR. Surprisingly, we revealed that valerate, hexanoate, and heptanoate with 5-7 carbons are more potent than 4-carbon butyrate in promoting LL-37 gene expression in both cell types. Free fatty acids with longer than 7 or shorter than 4 carbons showed only a marginal effect on LL-37 expression. Studies with a series of fatty acid derivatives with modifications in the aliphatic chain or carboxylic acid group yielded several analogs such as benzyl butyrate, trans-cinnamyl butyrate, glyceryl tributyrate, and phenethyl butyrate with a comparable LL-37-inducing activity to sodium butyrate. On the other hand, although reactive, the anhydride derivatives of short- and medium-chain fatty acids are as potent as their corresponding free acid forms in LL-37 induction. Thus, these newly identified free fatty acids and their analogs with a strong capacity to augment LL-37 synthesis may hold promise as immune boosting dietary supplements for antimicrobial therapy.
Collapse
Affiliation(s)
- Weiyu Jiang
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74048, USA
| | | | | | | | | | | |
Collapse
|
25
|
Zeng X, Sunkara LT, Jiang W, Bible M, Carter S, Ma X, Qiao S, Zhang G. Induction of porcine host defense peptide gene expression by short-chain fatty acids and their analogs. PLoS One 2013; 8:e72922. [PMID: 24023657 PMCID: PMC3758276 DOI: 10.1371/journal.pone.0072922] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/13/2013] [Indexed: 01/08/2023] Open
Abstract
Dietary modulation of the synthesis of endogenous host defense peptides (HDPs) represents a novel antimicrobial approach for disease control and prevention, particularly against antibiotic-resistant infections. However, HDP regulation by dietary compounds such as butyrate is species-dependent. To examine whether butyrate could induce HDP expression in pigs, we evaluated the expressions of a panel of porcine HDPs in IPEC-J2 intestinal epithelial cells, 3D4/31 macrophages, and primary monocytes in response to sodium butyrate treatment by real-time PCR. We revealed that butyrate is a potent inducer of multiple, but not all, HDP genes. Porcine β-defensin 2 (pBD2), pBD3, epididymis protein 2 splicing variant C (pEP2C), and protegrins were induced markedly in response to butyrate, whereas pBD1 expression remained largely unaltered in any cell type. Additionally, a comparison of the HDP-inducing efficacy among saturated free fatty acids of different aliphatic chain lengths revealed that fatty acids containing 3–8 carbons showed an obvious induction of HDP expression in IPEC-J2 cells, with butyrate being the most potent and long-chain fatty acids having only a marginal effect. We further investigated a panel of butyrate analogs for their efficacy in HDP induction, and found glyceryl tributyrate, benzyl butyrate, and 4-phenylbutyrate to be comparable with butyrate. Identification of butyrate and several analogs with a strong capacity to induce HDP gene expression in pigs provides attractive candidates for further evaluation of their potential as novel alternatives to antibiotics in augmenting innate immunity and disease resistance of pigs.
Collapse
Affiliation(s)
- Xiangfang Zeng
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Lakshmi T. Sunkara
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Weiyu Jiang
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Megan Bible
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Scott Carter
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, United States of America
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
26
|
DA SILVA JEFERSONG, PERDIGÃO CAMILAC, SPEZIALI NIVALDOL, BERALDO HELOISA. Chalcone-derived thiosemicarbazones and their zinc(II) and gallium(III) complexes: spectral studies and antimicrobial activity. J COORD CHEM 2013. [DOI: 10.1080/00958972.2012.757762] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- JEFERSON G. DA SILVA
- a Departamento de Química , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| | - CAMILA C.H. PERDIGÃO
- a Departamento de Química , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| | - NIVALDO L. SPEZIALI
- b Departamento de Física , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| | - HELOISA BERALDO
- a Departamento de Química , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| |
Collapse
|
27
|
Hiramatsu Y, Satho T, Irie K, Shiimura S, Okuno T, Sharmin T, Uyeda S, Fukumitsu Y, Nakashima Y, Miake F, Kashige N. Differences in TLR9-dependent inhibitory effects of H(2)O(2)-induced IL-8 secretion and NF-kappa B/I kappa B-alpha system activation by genomic DNA from five Lactobacillus species. Microbes Infect 2012. [PMID: 23182970 DOI: 10.1016/j.micinf.2012.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lactic acid bacteria (LAB) show anti-inflammatory effects, and their genomic DNA was identified as one of the anti-inflammatory components. Despite the differences in anti-inflammatory effects between live LAB dependent not only on genus but also species, this effect has not been compared at the genomic DNA level. We compared the anti-inflammatory effects of the genomic DNA from five Lactobacillus species-Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus gasseri, Lactobacillus plantarum, and Lactobacillus reuteri-using Caco-2 cells. To evaluate anti-inflammatory effects, decreases in H(2)O(2)-induced IL-8 secretion and inhibition of H(2)O(2)-induced NF-κB/IκB-α system activation were examined. All LAB genomic DNAs dose-dependently decreased H(2)O(2)-induced IL-8 secretion and inhibited H(2)O(2)-induced NF-κB/IκB-α system activation. Comparison of these effects between Lactobacillus species showed that the anti-inflammatory effects of L. acidophilus genomic DNA are lower than those of the other species. Furthermore, suppression of Toll-like receptor 9 (TLR9), a specific receptor of bacterial DNA, expression by RNAi abolished the decrease of H(2)O(2)-induced IL-8 secretion and inhibition of H(2)O(2)-induced NF-κB/IκB-α system activation by LAB genomic DNA. Our results demonstrated that the anti-inflammatory effects of genomic DNA differ between Lactobacillus species and TLR9 is one of the major pathways responsible for the anti-inflammatory effect of LAB genomic DNA.
Collapse
|
28
|
Chromek M, Arvidsson I, Karpman D. The antimicrobial peptide cathelicidin protects mice from Escherichia coli O157:H7-mediated disease. PLoS One 2012; 7:e46476. [PMID: 23077510 PMCID: PMC3471911 DOI: 10.1371/journal.pone.0046476] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 08/30/2012] [Indexed: 12/16/2022] Open
Abstract
This study investigated the role of the antimicrobial peptide cathelicidin in Escherichia coli O157:H7 infection and subsequent renal damage. Mouse and human cathelicidin, CRAMP and LL-37, respectively, killed E. coli O157:H7 in vitro. Intestines from healthy wild-type (129/SvJ) and cathelicidin-knock-out (Camp−/−) mice were investigated, showing that cathelicidin-deficient mice had a thinner colonic mucus layer compared with wild-type mice. Wild-type (n = 11) and cathelicidin-knock-out (n = 11) mice were inoculated with E. coli O157:H7. Cathelicidin-deficient animals exhibited higher fecal counts of E. coli O157:H7 and bacteria penetrated the mucus forming attaching-and-effacing lesions to a much higher extent than in wild-type animals. Cathelicidin knock-out mice developed symptoms (9/11) as well as anemia, thrombocytopenia and extensive renal tubular damage while all cathelicidin-producing mice remained asymptomatic with normal laboratory findings. When injected with Shiga toxin intraperitoneally, both murine strains developed the same degree of renal tubular damage and clinical disease indicating that differences in sensitivity to infection between the murine strains were related to the initial intestinal response. In conclusion, cathelicidin substantially influenced the antimicrobial barrier in the mouse colon mucosa. Cathelicidin deficiency lead to increased susceptibility to E. coli O157:H7 infection and subsequent renal damage. Administration of cathelicidin or stimulation of endogenous production may prove to be novel treatments for E. coli O157:H7-induced hemolytic uremic syndrome.
Collapse
Affiliation(s)
| | | | - Diana Karpman
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
- * E-mail:
| |
Collapse
|
29
|
|
30
|
Tripathi A, Saravanan S, Pattnaik S, Moorthi A, Partridge NC, Selvamurugan N. Bio-composite scaffolds containing chitosan/nano-hydroxyapatite/nano-copper-zinc for bone tissue engineering. Int J Biol Macromol 2011; 50:294-9. [PMID: 22123094 DOI: 10.1016/j.ijbiomac.2011.11.013] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 11/11/2011] [Accepted: 11/14/2011] [Indexed: 11/30/2022]
Abstract
The current study involves fabrication and characterization of bio-composite scaffolds containing chitosan (CS), nano-hydroxyapatite (nHAp) and Cu-Zn alloy nanoparticles (nCu-Zn) by freeze drying technique. The fabricated composite scaffolds (CS/nHAp and CS/nHAp/nCu-Zn) were characterized by SEM, EDX, XRD and FT-IR studies. The addition of nCu-Zn in the CS/nHAp scaffolds significantly increased swelling, decreased degradation, increased protein adsorption, and increased antibacterial activity. The CS/nHAp/nCu-Zn scaffolds had no toxicity towards rat osteoprogenitor cells. So the developed CS/nHAp/nCu-Zn scaffolds have advantageous and potential applications over the CS-nHAp scaffolds for bone tissue engineering.
Collapse
Affiliation(s)
- Anjali Tripathi
- Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur, Tamil Nadu, India
| | | | | | | | | | | |
Collapse
|
31
|
Saravanan S, Nethala S, Pattnaik S, Tripathi A, Moorthi A, Selvamurugan N. Preparation, characterization and antimicrobial activity of a bio-composite scaffold containing chitosan/nano-hydroxyapatite/nano-silver for bone tissue engineering. Int J Biol Macromol 2011; 49:188-93. [DOI: 10.1016/j.ijbiomac.2011.04.010] [Citation(s) in RCA: 185] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 04/15/2011] [Accepted: 04/18/2011] [Indexed: 10/18/2022]
|