115 results on '"Archer, Nathan K."'
Search Results
2. A novel rodent model of chronic spinal implant-associated infection
- Author
-
DeMourdant, Trevor, Rajkovic, Christian J., Tracz, Jovanna A., Perdomo-Pantoja, Alexander, Judy, Brendan F., Hernandez, Vaughn N., Lin, Jessica, Lazzari, Julianna L., Dikeman, Dustin A., Archer, Nathan K., Davis, Kimberly M., Gordon, Oren, and Witham, Timothy F.
- Published
- 2023
- Full Text
- View/download PDF
3. Cluster Analysis of Circulating Plasma Biomarkers in Prurigo Nodularis Reveals a Distinct Systemic Inflammatory Signature in African Americans
- Author
-
Sutaria, Nishadh, Alphonse, Martin Prince, Marani, Melika, Parthasarathy, Varsha, Deng, Junwen, Wongvibulsin, Shannon, Williams, Kyle, Roh, Youkyung Sophie, Choi, Justin, Bordeaux, Zachary, Pritchard, Thomas, Dillen, Carly, Semenov, Yevgeniy R., Kwatra, Madan M., Archer, Nathan K., Garza, Luis A., Dong, Xinzhong, Kang, Sewon, and Kwatra, Shawn G.
- Published
- 2022
- Full Text
- View/download PDF
4. IL-6R/Signal Transducer and Activator of Transcription 3 Signaling in Keratinocytes rather than in T Cells Induces Psoriasis-Like Dermatitis in Mice
- Author
-
Ravipati, Advaitaa, Nolan, Sabrina, Alphonse, Martin, Dikeman, Dustin, Youn, Christine, Wang, Yu, Orlando, Nicholas, Patrick, Garrett, Lee, Steven, Ortines, Roger V., Liu, Haiyun, Miller, Robert J., Dillen, Carly A., Marchitto, Mark, Cai, S. Sarah, Miller, Lloyd S., and Archer, Nathan K.
- Published
- 2022
- Full Text
- View/download PDF
5. Staphylococcus aureus Proteases: Orchestrators of Skin Inflammation.
- Author
-
Kline, Sabrina N., Saito, Yoshine, and Archer, Nathan K.
- Subjects
SOFT tissue infections ,SKIN inflammation ,ATOPIC dermatitis ,STAPHYLOCOCCUS aureus ,GRAM-positive bacteria - Abstract
Skin homeostasis relies on a delicate balance between host proteases and protease inhibitors along with those secreted from microbial communities, as disruption to this harmony contributes to the pathogenesis of inflammatory skin disorders, including atopic dermatitis and Netherton's syndrome. In addition to being a prominent cause of skin and soft tissue infections, the gram-positive bacterium Staphylococcus aureus is a key player in inflammatory skin conditions due to its array of 10 secreted proteases. Herein we review how S. aureus proteases augment the development of inflammation in skin disorders. These mechanisms include degradation of skin barrier integrity, immune dysregulation and pruritis, and impairment of host defenses. Delineating the diverse roles of S. aureus proteases has the potential to reveal novel therapeutic strategies, such as inhibitors of proteases or their cognate target, as well as neutralizing vaccines to alleviate the burden of inflammatory skin disorders in patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Prurigo Nodularis Is Characterized by Systemic and Cutaneous T Helper 22 Immune Polarization
- Author
-
Belzberg, Micah, Alphonse, Martin Prince, Brown, Isabelle, Williams, Kyle A., Khanna, Raveena, Ho, Byron, Wongvibulsin, Shannon, Pritchard, Thomas, Roh, Youkyung Sophie, Sutaria, Nishadh, Choi, Justin, Jedrych, Jaroslaw, Johnston, Andrew D., Sarkar, Kakali, Vasavda, Chirag, Meixiong, Jimmy, Dillen, Carly, Bondesgaard, Kent, Paolini, John F., Chen, Wei, Corcoran, David, Devos, Nicolas, Kwatra, Madan M., Chien, Anna L., Archer, Nathan K., Garza, Luis A., Dong, Xinzhong, Kang, Sewon, and Kwatra, Shawn G.
- Published
- 2021
- Full Text
- View/download PDF
7. Which Way Do We Go? Complex Interactions in Atopic Dermatitis Pathogenesis
- Author
-
Patrick, Garrett J., Archer, Nathan K., and Miller, Lloyd S.
- Published
- 2021
- Full Text
- View/download PDF
8. Research Techniques Made Simple: Mouse Bacterial Skin Infection Models for Immunity Research
- Author
-
Youn, Christine, Archer, Nathan K., and Miller, Lloyd S.
- Published
- 2020
- Full Text
- View/download PDF
9. Clonal Vγ6⁺Vδ4⁺ T cells promote IL-17–mediated immunity against Staphylococcus aureus skin infection
- Author
-
Marchitto, Mark C., Dillen, Carly A., Liu, Haiyun, Miller, Robert J., Archer, Nathan K., Ortines, Roger V., Alphonse, Martin P., Marusina, Alina I., Merleev, Alexander A., Wang, Yu, Pinsker, Bret L., Byrd, Angel S., Brown, Isabelle D., Ravipati, Advaitaa, Zhang, Emily, Cai, Shuting S., Limjunyawong, Nathachit, Dong, Xinzhong, Yeaman, Michael R., Simon, Scott I., Shen, Wei, Durum, Scott K., O’Brien, Rebecca L., Maverakis, Emanual, and Miller, Lloyd S.
- Published
- 2019
10. Tick extracellular vesicles enable arthropod feeding and promote distinct outcomes of bacterial infection
- Author
-
Oliva Chávez, Adela S., Wang, Xiaowei, Marnin, Liron, Archer, Nathan K., Hammond, Holly L., Carroll, Erin E. McClure, Shaw, Dana K., Tully, Brenden G., Buskirk, Amanda D., Ford, Shelby L., Butler, L. Rainer, Shahi, Preeti, Morozova, Kateryna, Clement, Cristina C., Lawres, Lauren, Neal, Anya J. O’, Mamoun, Choukri Ben, Mason, Kathleen L., Hobbs, Brandi E., Scoles, Glen A., Barry, Eileen M., Sonenshine, Daniel E., Pal, Utpal, Valenzuela, Jesus G., Sztein, Marcelo B., Pasetti, Marcela F., Levin, Michael L., Kotsyfakis, Michail, Jay, Steven M., Huntley, Jason F., Miller, Lloyd S., Santambrogio, Laura, and Pedra, Joao H. F.
- Published
- 2021
- Full Text
- View/download PDF
11. Injury, dysbiosis, and filaggrin deficiency drive skin inflammation through keratinocyte IL-1α release
- Author
-
Archer, Nathan K., Jo, Jay-Hyun, Lee, Steven K., Kim, Dongwon, Smith, Barbara, Ortines, Roger V., Wang, Yu, Marchitto, Mark C., Ravipati, Advaitaa, Cai, Shuting S., Dillen, Carly A., Liu, Haiyun, Miller, Robert J., Ashbaugh, Alyssa G., Uppal, Angad S., Oyoshi, Michiko K., Malhotra, Nidhi, Hoff, Sabine, Garza, Luis A., Kong, Heidi H., Segre, Julia A., Geha, Raif S., and Miller, Lloyd S.
- Published
- 2019
- Full Text
- View/download PDF
12. Dry and liquid formulations of IBT-V02, a novel multi-component toxoid vaccine, are effective against Staphylococcus aureus isolates from low-to-middle income countries.
- Author
-
Yu Wang, Mukherjee, Ipsita, Venkatasubramaniam, Arundhathi, Dikeman, Dustin, Orlando, Nicholas, Jing Zhang, Ortines, Roger, Mednikov, Mark, Sherchand, Shardulendra P., Kanipakala, Tulasikumari, Thao Le, Shukla, Sanjay, Ketner, Mark, Adhikari, Rajan P., Karauzum, Hatice, Aman, M. Javad, and Archer, Nathan K.
- Subjects
STAPHYLOCOCCUS aureus ,SOFT tissue infections ,SURGICAL site infections ,OSTEOMYELITIS ,METHICILLIN-resistant staphylococcus aureus ,RABBIT diseases ,VACCINES - Abstract
Staphylococcus aureus is the leading cause of skin and soft tissue infections (SSTIs) in the U.S. as well as more serious invasive diseases, including bacteremia, sepsis, endocarditis, surgical site infections, osteomyelitis, and pneumonia. These infections are exacerbated by the emergence of antibiotic-resistant clinical isolates such as methicillin-resistant S. aureus (MRSA), highlighting the need for alternatives to antibiotics to treat bacterial infections. We have previously developed a multi-component toxoid vaccine (IBT-V02) in a liquid formulation with efficacy against multiple strains of Staphylococcus aureus prevalent in the industrialized world. However, liquid vaccine formulations are not compatible with the paucity of cold chain storage infrastructure in many low-to-middle income countries (LMICs). Furthermore, whether our IBT-V02 vaccine formulations are protective against S. aureus isolates from LMICs is unknown. To overcome these limitations, we developed lyophilized and spray freeze-dried formulations of IBT-V02 vaccine and demonstrated that both formulations had comparable biophysical attributes as the liquid formulation, including similar levels of toxin neutralizing antibodies and protective efficacy against MRSA infections in murine and rabbit models. To enhance the relevancy of our findings, we then performed a multi-dimensional screen of 83 S. aureus clinical isolates from LMICs (e.g., Democratic Republic of Congo, Palestine, and Cambodia) to rationally down-select strains to test in our in vivo models based on broad expression of IBT-V02 targets (i.e., pore-forming toxins and superantigens). IBT-V02 polyclonal antisera effectively neutralized toxins produced by the S. aureus clinical isolates from LMICs. Notably, the lyophilized IBT-V02 formulation exhibited significant in vivo efficacy in various preclinical infection models against the S. aureus clinical isolates from LMICs, which was comparable to our liquid formulation. Collectively, our findings suggested that lyophilization is an effective alternative to liquid vaccine formulations of our IBTV02 vaccine against S. aureus infections, which has important implications for protection from S. aureus isolates from LMICs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. The 2023 Orthopaedic Research Society's International Consensus Meeting on musculoskeletal infection: Summary from the host immunity section.
- Author
-
Schwarz, Edward M., Archer, Nathan K., Atkins, Gerald J., Bentley, Karen L. de Mesy, Botros, Mina, Cassat, James E., Chisari, Emanuele, Coraça‐Huber, Débora C., Daiss, John L., Gill, Steven R., Goodman, Stuart B., Harro, Janette, Hernandez, Christopher J., Ivashkiv, Lionel B., Kates, Stephen L., Marques, Cláudia N. H., Masters, Elysia A., Muthukrishnan, Gowrishankar, Owen, John R., and Raafat, Dina
- Subjects
- *
PROSTHESIS-related infections , *INTERNET voting , *IMMUNITY , *INTERNET access , *VACCINE effectiveness - Abstract
Musculoskeletal infections (MSKI), which are a major problem in orthopedics, occur when the pathogen eludes or overwhelms the host immune system. While effective vaccines and immunotherapies to prevent and treat MSKI should be possible, fundamental knowledge gaps in our understanding of protective, nonprotective, and pathogenic host immunity are prohibitive. We also lack critical knowledge of how host immunity is affected by the microbiome, implants, prior infection, nutrition, antibiotics, and concomitant therapies, autoimmunity, and other comorbidities. To define our current knowledge of these critical topics, a Host Immunity Section of the 2023 Orthopaedic Research Society MSKI International Consensus Meeting (ICM) proposed 78 questions. Systematic reviews were performed on 15 of these questions, upon which recommendations with level of evidence were voted on by the 72 ICM delegates, and another 12 questions were voted on with a recommendation of "Unknown" without systematic reviews. Two questions were transferred to another ICM Section, and the other 45 were tabled for future consideration due to limitations of available human resources. Here we report the results of the voting with internet access to the questions, recommendations, and rationale from the systematic reviews. Eighteen questions received a consensus vote of ≥90%, while nine recommendations failed to achieve this threshold. Commentary on why consensus was not achieved on these questions and potential ways forward are provided to stimulate specific funding mechanisms and research on these critical MSKI host defense questions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Bactericidal Efficacy of the Combination of Maresin-like Proresolving Mediators and Carbenicillin Action on Biofilm-Forming Burn Trauma Infection-Related Bacteria.
- Author
-
Thamizhchelvan, Anbu Mozhi, Masoud, Abdul-Razak, Su, Shanchun, Lu, Yan, Peng, Hongying, Kobayashi, Yuichi, Wang, Yu, Archer, Nathan K., and Hong, Song
- Subjects
UNSATURATED fatty acids ,GRAM-negative bacteria ,BACTERIA ,DOCOSAHEXAENOIC acid ,GRAM-positive bacteria ,MARL ,MASTITIS - Abstract
Biofilm-associated bacterial infections are the major reason for treatment failure in many diseases including burn trauma infections. Uncontrolled inflammation induced by bacteria leads to materiality, tissue damage, and chronic diseases. Specialized proresolving mediators (SPMs), including maresin-like lipid mediators (MarLs), are enzymatically biosynthesized from omega-3 essential long-chain polyunsaturated fatty acids, especially docosahexaenoic acid (DHA), by macrophages and other leukocytes. SPMs exhibit strong inflammation-resolving activities, especially inflammation provoked by bacterial infection. In this study, we explored the potential direct inhibitory activities of three MarLs on Gram-positive (Staphylococcus aureus) and Gram-negative (Pseudomonas aeruginosa and Escherichia coli) bacteria in their biofilms that are leading bacteria in burn trauma-related infections. We also examined the effects of MarLs on the bactericidal activities of a typical broad-spectrum antibiotic, carbenicillin (carb), on these bacteria in their preformed biofilms. The results revealed that MarLs combined with carbenicillin can inhibit the survival of Gram-positive and Gram-negative bacteria in their biofilms although MarLs alone did not exhibit bactericidal activity. Thus, our findings suggest that the combination of MarLs and carbenicillin can lower the antibiotic requirements to kill the bacteria in preformed biofilms. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Comparative intravital imaging of human and rodent malaria sporozoites reveals the skin is not a species‐specific barrier
- Author
-
Hopp, Christine S, Kanatani, Sachie, Archer, Nathan K, Miller, Robert J, Liu, Haiyun, Chiou, Kevin K, Miller, Lloyd S, and Sinnis, Photini
- Published
- 2021
- Full Text
- View/download PDF
16. Mouse model of hematogenous implant-related Staphylococcus aureus biofilm infection reveals therapeutic targets
- Author
-
Wang, Yu, Cheng, Lily I., Helfer, David R., Ashbaugh, Alyssa G., Miller, Robert J., Tzomides, Alexander J., Thompson, John M., Ortines, Roger V., Tsai, Andrew S., Liu, Haiyun, Dillen, Carly A., Archer, Nathan K., Cohen, Taylor S., Tkaczyk, Christine, Stover, C. Kendall, Sellman, Bret R., and Miller, Lloyd S.
- Published
- 2017
17. Neutrophil extracellular trap-associated RNA and LL37 enable self-amplifying inflammation in psoriasis
- Author
-
Herster, Franziska, Bittner, Zsofia, Archer, Nathan K., Dickhöfer, Sabine, Eisel, David, Eigenbrod, Tatjana, Knorpp, Thomas, Schneiderhan-Marra, Nicole, Löffler, Markus W., Kalbacher, Hubert, Vierbuchen, Tim, Heine, Holger, Miller, Lloyd S., Hartl, Dominik, Freund, Lukas, Schäkel, Knut, Heister, Martin, Ghoreschi, Kamran, and Weber, Alexander N. R.
- Published
- 2020
- Full Text
- View/download PDF
18. Staphylococcus aureus proteases trigger eosinophil-mediated skin inflammation.
- Author
-
Kline, Sabrina N., Orlando, Nicholas A., Lee, Alex J., Meng-Jen Wu, Jing Zhang, Youn, Christine, Feller, Laine E., Pontaza, Cristina, Dikeman, Dustin, Limjunyawong, Nathachit, Williams, Kaitlin L., Yu Wang, Cihakova, Daniela, Jacobsen, Elizabeth A., Durum, Scott K., Garza, Luis A., Xinzhong Dong, and Archer, Nathan K.
- Subjects
SKIN inflammation ,STAPHYLOCOCCUS aureus ,BULLOUS pemphigoid ,PROTEOLYTIC enzymes ,ATOPIC dermatitis - Abstract
Staphylococcus aureus skin colonization and eosinophil infiltration are associated with many inflammatory skin disorders, including atopic dermatitis, bullous pemphigoid, Netherton’s syndrome, and prurigo nodularis. However, whether there is a relationship between S. aureus and eosinophils and how this interaction influences skin inflammation is largely undefined. We show in a preclinical mouse model that S. aureus epicutaneous exposure induced eosinophil-recruiting chemokines and eosinophil infiltration into the skin. Remarkably, we found that eosinophils had a comparable contribution to the skin inflammation as T cells, in a manner dependent on eosinophil-derived IL-17A and IL-17F production. Importantly, IL-36R signaling induced CCL7-mediated eosinophil recruitment to the inflamed skin. Last, S. aureus proteases induced IL-36α expression in keratinocytes, which promoted infiltration of IL-17- producing eosinophils. Collectively, we uncovered a mechanism for S. aureus proteases to trigger eosinophil-mediated skin inflammation, which has implications in the pathogenesis of inflammatory skin diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Clonally expanded [gamma][delta] T cells protect against Staphylococcus aureus skin reinfection
- Author
-
Dillen, Carly A., Pinsker, Bret L., Marusina, Alina I., Merleev, Alexander A., Farber, Orly N., Liu, Haiyun, Archer, Nathan K., Lee, Da.B., Wang, Yu, Ortines, Roger V., Lee, Steven K., Marchitto, Mark C., Cai, Shuting S., Ashbaugh, Alyssa G., May, Larissa S., Holland, Steven M., Freeman, Alexandra F., Miller, Loren G., Yeaman, Michael R., Simon, Scott I., Milner, Joshua D., Maverakis, Emanual, and Miller, Lloyd S.
- Subjects
Genotypes -- Health aspects ,Staphylococcus aureus infections -- Genetic aspects -- Development and progression -- Care and treatment ,Infectious skin diseases -- Genetic aspects -- Development and progression -- Care and treatment ,T cells -- Genetic aspects -- Health aspects ,Health care industry - Abstract
The mechanisms that mediate durable protection against Staphylococcus aureus skin reinfections are unclear, as recurrences are common despite high antibody titers and memory T cells. Here, we developed a mouse model of S. aureus skin reinfection to investigate protective memory responses. In contrast with WT mice, IL-1[beta]-deficient mice exhibited poor neutrophil recruitment and bacterial clearance during primary infection that was rescued during secondary S. aureus challenge. The [gamma][delta] T cells from skin-draining LNs utilized compensatory T cell-intrinsic TLR2/MyD88 signaling to mediate rescue by trafficking and producing TNF and IFN-[gamma], which restored neutrophil recruitment and promoted bacterial clearance. RNA-sequencing (RNA-seq) of the LNs revealed a clonotypic S. aureus-induced [gamma][delta] T cell expansion with a complementarity-determining region 3 (CDR3) aa sequence identical to that of invariant V[gamma][5.sup.+] dendritic epidermal T cells. However, this T cell receptor [gamma] (TRG) aa sequence of the dominant CDR3 sequence was generated from multiple gene rearrangements of TRGV5 and TRGV6, indicating clonotypic expansion. TNF- and IFN-[gamma]- producing [gamma][delta] T cells were also expanded in peripheral blood of IRAK4-deficient humans no longer predisposed to S. aureus skin infections. Thus, clonally expanded [gamma][delta] T cells represent a mechanism for long-lasting immunity against recurrent S. aureus skin infections., Introduction Staphylococcus aureus is a Gram-positive bacterium that is the most common cause of skin infections in humans and is also an important cause of invasive and life-threatening infections, such [...]
- Published
- 2018
- Full Text
- View/download PDF
20. Collaborative Interferon-γ and Interleukin-17 Signaling Protects the Oral Mucosa from Staphylococcus aureus
- Author
-
Barin, Jobert G., Talor, Monica V., Schaub, Julie A., Diny, Nicola L., Hou, Xuezhou, Hoyer, Matthew, Archer, Nathan K., Gebremariam, Elizabeth S., Davis, Meghan F., Miller, Lloyd S., Rose, Noel R., and Čiháková, Daniela
- Published
- 2016
- Full Text
- View/download PDF
21. In Vivo Bioluminescence Imaging in a Rabbit Model of Orthopaedic Implant-Associated Infection to Monitor Efficacy of an Antibiotic-Releasing Coating
- Author
-
Miller, Robert J., Thompson, John M., Zheng, Jesse, Marchitto, Mark C., Archer, Nathan K., Pinsker, Bret L., Ortines, Roger V., Jiang, Xuesong, Martin, Russell A., Brown, Isabelle D., Wang, Yu, Sterling, Robert S., Mao, Hai-Quan, and Miller, Lloyd S.
- Published
- 2019
- Full Text
- View/download PDF
22. Development of a Staphylococcus aureus reporter strain with click beetle red luciferase for enhanced in vivo imaging of experimental bacteremia and mixed infections
- Author
-
Miller, Robert J., Crosby, Heidi A., Schilcher, Katrin, Wang, Yu, Ortines, Roger V., Mazhar, Momina, Dikeman, Dustin A., Pinsker, Bret L., Brown, Isabelle D., Joyce, Daniel P., Zhang, Jeffrey, Archer, Nathan K., Liu, Haiyun, Alphonse, Martin P., Czupryna, Julie, Anderson, William R., Bernthal, Nicholas M., Fortuno-Miranda, Lea, Bulte, Jeff W. M., Francis, Kevin P., Horswill, Alexander R., and Miller, Lloyd S.
- Published
- 2019
- Full Text
- View/download PDF
23. Noncoding dsRNA induces retinoic acid synthesis to stimulate hair follicle regeneration via TLR3
- Author
-
Kim, Dongwon, Chen, Ruosi, Sheu, Mary, Kim, Noori, Kim, Sooah, Islam, Nasif, Wier, Eric M., Wang, Gaofeng, Li, Ang, Park, Angela, Son, Wooyang, Evans, Benjamin, Yu, Victoria, Prizmic, Vicky P., Oh, Eugene, Wang, Zixiao, Yu, Jianshi, Huang, Weiliang, Archer, Nathan K., Hu, Zhiqi, Clemetson, Nashay, Nelson, Amanda M., Chien, Anna, Okoye, Ginette A., Miller, Lloyd S., Ghiaur, Gabriel, Kang, Sewon, Jones, Jace W., Kane, Maureen A., and Garza, Luis A.
- Published
- 2019
- Full Text
- View/download PDF
24. Clonally expanded γδ T cells protect against Staphylococcus aureus skin reinfection
- Author
-
Dillen, Carly A., Pinsker, Bret L., Marusina, Alina I., Merleev, Alexander A., Farber, Orly N., Liu, Haiyun, Archer, Nathan K., Lee, Da B., Wang, Yu, Ortines, Roger V., Lee, Steven K., Marchitto, Mark C., Cai, Shuting S., Ashbaugh, Alyssa G., May, Larissa S., Holland, Steven M., Freeman, Alexandra F., Miller, Loren G., Yeaman, Michael R., Simon, Scott I., Milner, Joshua D., Maverakis, Emanual, and Miller, Lloyd S.
- Published
- 2018
- Full Text
- View/download PDF
25. γδ T cell-intrinsic IL-1R promotes survival during Staphylococcus aureus bacteremia.
- Author
-
Yu Wang, Ahmadi, Michael Z., Dikeman, Dustin A., Youn, Christine, and Archer, Nathan K.
- Subjects
BACTEREMIA ,STAPHYLOCOCCUS aureus ,T cells ,ENDOTHELIAL cells ,CELL survival - Abstract
Staphylococcus aureus is a leading cause of bacteremia, further complicated by the emergence of antibiotic-resistant strains such as methicillin-resistant S. aureus (MRSA). A better understanding of host defense mechanisms is needed for the development of host-directed therapies as an alternative approach to antibiotics. The levels of IL-1, IL-17, and TNF-α cytokines in circulation have been associated with predictive outcomes in patients with S. aureus bacteremia. However, their causative role in survival and the cell types involved in these responses during bacteremia is not entirely clear. Using a mouse model of S. aureus bacteremia, we demonstrated that IL-17A/F and TNF-α had no significant impact on survival, whereas IL-1R signaling was critical for survival during S. aureus bacteremia. Furthermore, we identified that T cells, but not neutrophils, monocytes/macrophages, or endothelial cells were the crucial cell type for IL-1R-mediated survival against S. aureus bacteremia. Finally, we determined that the expression of IL-1R on gd T cell, but not CD4
+ or CD8+ T cells was responsible for survival against the S. aureus bacteremia. Taken together, we uncovered a role for IL-1R, but not IL-17A/F and TNF-α in protection against S. aureus bacteremia. Importantly, γδ T cell-intrinsic expression of IL-1R was crucial for survival, but not on other immune cells or endothelial cells. These findings reveal potential cellular and immunological targets for host-directed therapies for improved outcomes against S. aureus bacteremia. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
26. Cutaneous Transcriptomics Identifies Fibroproliferative and Neurovascular Gene Dysregulation in Prurigo Nodularis Compared with Psoriasis and Atopic Dermatitis
- Author
-
Sutaria, Nishadh, Alphonse, Martin Prince, Roh, Youkyung S., Choi, Justin, Parthasarathy, Varsha, Deng, Junwen, Bordeaux, Zachary A., Taylor, Matthew T., Pritchard, Thomas, Kim, Noori, Aguh, Crystal, Semenov, Yevgeniy R., Archer, Nathan K., Garza, Luis A., Kang, Sewon, and Kwatra, Shawn G.
- Published
- 2022
- Full Text
- View/download PDF
27. Crisaborole efficacy in murine models of skin inflammation and Staphylococcus aureus infection.
- Author
-
Youn, Christine, Dikeman, Dustin A., Chang, Evelyn, Liu, Haiyun, Nolan, Sabrina J., Alphonse, Martin P., Joyce, Daniel P., Liu, Qi, Meixiong, James, Dong, Xinzhong, Miller, Lloyd S., and Archer, Nathan K.
- Subjects
STAPHYLOCOCCUS aureus infections ,SKIN inflammation ,ITCHING ,BACTERIAL colonies ,ATOPIC dermatitis ,SKIN diseases - Abstract
Phosphodiesterase 4 (PDE4) is highly expressed in keratinocytes and immune cells and promotes pro‐inflammatory responses upon activation. The activity of PDE4 has been attributed to various inflammatory conditions, leading to the development and approval of PDE4 inhibitors as host‐directed therapeutics in humans. For example, the topical PDE4 inhibitor, crisaborole, is approved for the treatment of mild‐to‐moderate atopic dermatitis and has shown efficacy in patients with psoriasis. However, the role of crisaborole in regulating the immunopathogenesis of inflammatory skin diseases and infection is not entirely known. Therefore, we evaluated the effects of crisaborole in multiple mouse models, including psoriasis‐like dermatitis, AD‐like skin inflammation with and without filaggrin mutations, and Staphylococcus aureus skin infection. We discovered that crisaborole dampens myeloid cells and itch in the skin during psoriasis‐like dermatitis. Furthermore, crisaborole was effective in reducing skin inflammation in the context of filaggrin deficiency. Importantly, crisaborole reduced S. aureus skin colonization during AD‐like skin inflammation. However, crisaborole was not efficacious in treating S. aureus skin infections, even as adjunctive therapy to antibiotics. Taken together, we found that crisaborole reduced itch during psoriasis‐like dermatitis and decreased S. aureus skin colonization upon AD‐like skin inflammation, which act as additional mechanisms by which crisaborole dampens the immunopathogenesis in mouse models of inflammatory skin diseases. Further examination is warranted to translate these preclinical findings to human disease. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
28. Staphylococcus aureus adaptive evolution: Recent insights on how immune evasion, immunometabolic subversion and host genetics impact vaccine development.
- Author
-
Tania Wong Fok Lung, Chan, Liana C., Prince, Alice, Yeaman, Michael R., Archer, Nathan K., Javad Aman, M., and Proctor, Richard A.
- Subjects
VACCINE development ,GENETICS ,HUMAN genetics ,VACCINE effectiveness ,TECHNOLOGICAL innovations - Abstract
Despite meritorious attempts, a S. aureus vaccine that prevents infection or mitigates severity has not yet achieved efficacy endpoints in prospective, randomized clinical trials. This experience underscores the complexity of host- S. aureus interactions, which appear to be greater than many other bacterial pathogens against which successful vaccines have been developed. It is increasingly evident that S. aureus employs strategic countermeasures to evade or exploit human immune responses. From entering host cells to persist in stealthy intracellular reservoirs, to sensing the environmental milieu and leveraging bacterial or host metabolic products to reprogram host immune responses, S. aureus poses considerable challenges for the development of effective vaccines. The fact that this pathogen causes distinct types of infections and can undergo transient genetic, transcriptional or metabolic adaptations in vivo that do not occur in vitro compounds challenges in vaccine development. Notably, themetabolic versatility of both bacterial and host immune cells as they compete for available substrates within specific tissues inevitably impacts the variable repertoire of gene products that may ormay not be vaccine antigens. In this respect, S. aureus has chameleon phenotypes that have alluded vaccine strategies thus far. Nonetheless, a number of recent studies have also revealed important new insights into pathogenesis vulnerabilities of S. aureus. A more detailed understanding of host protective immune defenses versus S. aureus adaptive immune evasion mechanisms may offer breakthroughs in the development of effective vaccines, but at present this goal remains a very high bar. Coupled with the recent advances in human genetics and epigenetics, newer vaccine technologies may enable such a goal. If so, future vaccines that protect against or mitigate the severity of S. aureus infections are likely to emerge at the intersection of precision and personalized medicine. For now, the development of S. aureus vaccines or alternative therapies that reduce mortality and morbidity must continue to be pursued. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
29. From top to bottom: Staphylococci in atopic dermatitis.
- Author
-
Nolan, Sabrina J. and Archer, Nathan K.
- Abstract
In this issue of Cell Host & Microbe , Kashaf et al. and Key et al. examine isolates of Staphylococcus aureus among individuals with atopic dermatitis, revealing insights into evolution, antibiotic resistance, transmission mechanisms, skin colonization, and virulence factors. These findings further our understanding of disease pathogenesis and potential treatments. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
30. CCR2 contributes to host defense against Staphylococcus aureus orthopedic implant‐associated infections in mice.
- Author
-
Wang, Yu, Dikeman, Dustin, Zhang, Jeffrey, Ackerman, Nicole, Kim, Sophia, Alphonse, Martin P., Ortines, Roger V., Liu, Haiyun, Joyce, Daniel P., Dillen, Carly A., Thompson, John M., Thomas, Abigail A., Plaut, Roger D., Miller, Lloyd S., and Archer, Nathan K.
- Subjects
CHEMOKINE receptors ,STAPHYLOCOCCUS aureus ,LYSIS ,MYELOID cells ,MICE - Abstract
C‐C motif chemokine receptor 2 (CCR2) is an important mediator of myeloid cell chemotaxis during inflammation and infection. Myeloid cells such as monocytes, macrophages, and neutrophils contribute to host defense during orthopedic implant‐associated infections (OIAI), but whether CCR2‐mediated chemotaxis is involved remains unclear. Therefore, a Staphylococcus aureus OIAI model was performed by surgically placing an orthopedic‐grade titanium implant and inoculating a bioluminescent S. aureus strain in knee joints of wildtype (wt) and CCR2‐deficient mice. In vivo bioluminescent signals significantly increased in CCR2‐deficient mice compared with wt mice at later time points (Days 14–28), which was confirmed with ex vivo colony‐forming unit enumeration. S. aureus γ‐hemolysin utilizes CCR2 to induce host cell lysis. However, there were no differences in bacterial burden when the OIAI model was performed with a parental versus a mutant γ‐hemolysin‐deficient S. aureus strain, indicating that the protection was mediated by the host cell function of CCR2 rather than γ‐hemolysin virulence. Although CCR2‐deficient and wt mice had similar cellular infiltrates in the infected joint tissue, CCR2‐deficient mice had reduced myeloid cells and γδ T cells in the draining lymph nodes. Taken together, CCR2 contributed to host defense at later time points during an OIAI by increasing immune cell infiltrates in the draining lymph nodes, which likely contained the infection and prevented invasive spread. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
31. Dynamic PET-facilitated modeling and high-dose rifampin regimens for Staphylococcus aureus orthopedic implant–associated infections.
- Author
-
Gordon, Oren, Lee, Donald E., Liu, Bessie, Langevin, Brooke, Ordonez, Alvaro A., Dikeman, Dustin A., Shafiq, Babar, Thompson, John M., Sponseller, Paul D., Flavahan, Kelly, Lodge, Martin A., Rowe, Steven P., Dannals, Robert F., Ruiz-Bedoya, Camilo A., Read, Timothy D., Peloquin, Charles A., Archer, Nathan K., Miller, Lloyd S., Davis, Kimberly M., and Gobburu, Jogarao V. S.
- Subjects
RIFAMPIN ,POSITRON emission tomography ,LABORATORY mice ,BACTERIAL mutation ,DYNAMIC models - Abstract
To the bone: Combination treatment with rifampin is recommend for Staphylococcus aureus orthopedic implant–associated infections yet requires prolonged treatment duration and has limited bone penetration. Gordon et al. used imaging and modeling to assess
11 C-rifampin concentration-time profiles in bone tissue in patients and a mouse model of implant-associated infection. The concentration of rifampin in bone was subtherapeutic when administered according to current dosing regimens, whereas higher dosing increased bacterial killing. Shorter-duration high-dose rifampin combination treatment was as effective as standard dose treatment in mice and was associated with reduced rifampin resistance mutations in bacteria, suggesting that high-dose regimens may improve outcomes. Staphylococcus aureus is a major human pathogen causing serious implant–associated infections. Combination treatment with rifampin (10 to 15 mg/kg per day), which has dose-dependent activity, is recommended to treat S. aureus orthopedic implant–associated infections. Rifampin, however, has limited bone penetration. Here, dynamic11 C-rifampin positron emission tomography (PET) performed in prospectively enrolled patients with confirmed S. aureus bone infection (n = 3) or without orthopedic infection (n = 12) demonstrated bone/plasma area under the concentration-time curve ratio of 0.14 (interquartile range, 0.09 to 0.19), exposures lower than previously thought. PET-based pharmacokinetic modeling predicted rifampin concentration-time profiles in bone and facilitated studies in a mouse model of S. aureus orthopedic implant infection. Administration of high-dose rifampin (human equipotent to 35 mg/kg per day) substantially increased bone concentrations (2 mg/liter versus <0.2 mg/liter with standard dosing) in mice and achieved higher bacterial killing and biofilm disruption. Treatment for 4 weeks with high-dose rifampin and vancomycin was noninferior to the recommended 6-week treatment of standard-dose rifampin with vancomycin in mice (risk difference, −6.7% favoring high-dose rifampin regimen). High-dose rifampin treatment ameliorated antimicrobial resistance (0% versus 38%; P = 0.04) and mitigated adverse bone remodeling (P < 0.01). Last, whole-genome sequencing demonstrated that administration of high-dose rifampin in mice reduced selection of bacterial mutations conferring rifampin resistance (rpoB) and mutations in genes potentially linked to persistence. These data suggest that administration of high-dose rifampin is necessary to achieve optimal bone concentrations, which could shorten and improve treatments for S. aureus orthopedic implant infections. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
32. Platelets Aggregate With Neutrophils and Promote Skin Pathology in Psoriasis
- Author
-
Herster, Franziska, Bittner, Zsofia, Codrea, Marius Cosmin, Archer, Nathan K., Heister, Martin, Löffler, Markus W., Heumos, Simon, Wegner, Joanna, Businger, Ramona, Schindler, Michael, Stegner, David, Schäkel, Knut, Grabbe, Stephan, Ghoreschi, Kamran, Miller, Lloyd S., and Weber, Alexander N. R.
- Subjects
platelet ,Adult ,Blood Platelets ,Imiquimod ,Platelet Aggregation ,Neutrophils ,Platelet Count ,Immunology ,neutrophil ,hemic and immune systems ,platelet-neutrophil complexes (PNCs) ,psoriasis ,Platelet Activation ,Mice, Inbred C57BL ,Mice ,Animals ,Humans ,Original Research ,Skin - Abstract
Psoriasis is a frequent systemic inflammatory autoimmune disease characterized primarily by skin lesions with massive infiltration of leukocytes, but frequently also presents with cardiovascular comorbidities. Especially polymorphonuclear neutrophils (PMNs) abundantly infiltrate psoriatic skin but the cues that prompt PMNs to home to the skin are not well-defined. To identify PMN surface receptors that may explain PMN skin homing in psoriasis patients, we screened 332 surface antigens on primary human blood PMNs from healthy donors and psoriasis patients. We identified platelet surface antigens as a defining feature of psoriasis PMNs, due to a significantly increased aggregation of neutrophils and platelets in the blood of psoriasis patients. Similarly, in the imiquimod-induced experimental in vivo mouse model of psoriasis, disease induction promoted PMN-platelet aggregate formation. In psoriasis patients, disease incidence directly correlated with blood platelet counts and platelets were detected in direct contact with PMNs in psoriatic but not healthy skin. Importantly, depletion of circulating platelets in mice in vivo ameliorated disease severity significantly, indicating that both PMNs and platelets may be relevant for psoriasis pathology and disease severity.
- Published
- 2019
33. Neutrophil extracellular traps impair regeneration.
- Author
-
Wier, Eric, Asada, Mayumi, Wang, Gaofeng, Alphonse, Martin P., Li, Ang, Hintelmann, Chase, Sweren, Evan, Youn, Christine, Pielstick, Brittany, Ortines, Roger, Lyu, Chenyi, Daskam, Maria, Miller, Lloyd S., Archer, Nathan K., and Garza, Luis A.
- Subjects
EMBRYOLOGY ,HAIR follicles ,NEUTROPHILS ,RNA sequencing ,DOUBLE-stranded RNA ,HEALING - Abstract
Fibrosis is a major health burden across diseases and organs. To remedy this, we study wound‐induced hair follicle neogenesis (WIHN) as a model of non‐fibrotic healing that recapitulates embryogenesis for de novo hair follicle morphogenesis after wounding. We previously demonstrated that TLR3 promotes WIHN through binding wound‐associated dsRNA, the source of which is still unclear. Here, we find that multiple distinct contexts of high WIHN all show a strong neutrophil signature. Given the correlation between neutrophil infiltration and endogenous dsRNA release, we hypothesized that neutrophil extracellular traps (NETs) likely release nuclear spliceosomal U1 dsRNA and modulate WIHN. However, rather than enhance regeneration, we find mature neutrophils inhibit WIHN such that mice with mature neutrophil depletion exhibit higher WIHN. Similarly, Pad4 null mice, which are defective in NET production, show augmented WIHN. Finally, using single‐cell RNA sequencing, we identify a dramatic increase in mature and activated neutrophils in the wound beds of low regenerating Tlr3−/− mice. Taken together, these results demonstrate that although mature neutrophils are stimulated by a common pro‐regenerative cue, their presence and NETs hinder regeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
34. Pan-caspase inhibition as a potential host-directed immunotherapy against MRSA and other bacterial skin infections.
- Author
-
Alphonse, Martin P., Rubens, Jessica H., Ortines, Roger V., Orlando, Nicholas A., Patel, Aman M., Dikeman, Dustin, Wang, Yu, Vuong, Ivan, Joyce, Daniel P., Zhang, Jeffrey, Mumtaz, Mohammed, Liu, Haiyun, Liu, Qi, Youn, Christine, Patrick, Garrett J., Ravipati, Advaitaa, Miller, Robert J., Archer, Nathan K., and Miller, Lloyd S.
- Abstract
Anti-infection immunotherapy: Antibiotic-resistant bacteria such as methicillin-resistant Staphylococcus aureus (MRSA) are increasingly prevalent and difficult to treat. Alphonse et al. show that a pan-caspase inhibitor called Q-VD-OPH functioned as an effective immunotherapy in mouse models of community-acquired MRSA, Streptococcus pyogenes, and Pseudomonas aeruginosa skin infections. Q-VD-OPH reduced apoptosis in neutrophils and monocytes and increased necroptosis in macrophages, thereby increasing TNF production and infection clearance in mice. This work suggests a potential strategy to target bacterial infections without requiring the use of antibiotics. Staphylococcus aureus causes most skin infections in humans, and the emergence of methicillin-resistant S. aureus (MRSA) strains is a serious public health threat. There is an urgent clinical need for nonantibiotic immunotherapies to treat MRSA infections and prevent the spread of antibiotic resistance. Here, we investigated the pan-caspase inhibitor quinoline–valine–aspartic acid–difluorophenoxymethyl ketone (Q-VD-OPH) for efficacy against MRSA skin infection in mice. A single systemic dose of Q-VD-OPH decreased skin lesion sizes and reduced bacterial burden compared with vehicle-treated or untreated mice. Although Q-VD-OPH inhibited inflammasome-dependent apoptosis-associated speck-like protein containing caspase activation and recruitment domain (ASC) speck formation and caspase-1–mediated interleukin-1β (IL-1β) production, Q-VD-OPH maintained efficacy in mice deficient in IL-1β, ASC, caspase-1, caspase-11, or gasdermin D. Thus, Q-VD-OPH efficacy was independent of inflammasome-mediated pyroptosis. Rather, Q-VD-OPH reduced apoptosis of monocytes and neutrophils. Moreover, Q-VD-OPH enhanced necroptosis of macrophages with concomitant increases in serum TNF and TNF-producing neutrophils, monocytes/macrophages, and neutrophils in the infected skin. Consistent with this, Q-VD-OPH lacked efficacy in mice deficient in TNF (with associated reduced neutrophil influx and necroptosis), in mice deficient in TNF/IL-1R and anti-TNF antibody-treated WT mice. In vitro studies revealed that combined caspase-3, caspase-8, and caspase-9 inhibition reduced apoptosis, and combined caspase-1, caspase-8, and caspase-11 inhibition increased TNF, suggesting a mechanism for Q-VD-OPH efficacy in vivo. Last, Q-VD-OPH also had a therapeutic effect against Streptococcus pyogenes and Pseudomonas aeruginosa skin infections in mice. Collectively, pan-caspase inhibition represents a potential host-directed immunotherapy against MRSA and other bacterial skin infections. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
35. Pushing the Envelope in Psoriasis: Late Cornified Envelope Proteins Possess Antimicrobial Activity
- Author
-
Archer, Nathan K., Dilolli, Migena N., and Miller, Lloyd S.
- Published
- 2017
- Full Text
- View/download PDF
36. Interleukin‐1β and tumor necrosis factor are essential in controlling an experimental orthopedic implant‐associated infection.
- Author
-
Wang, Yu, Ashbaugh, Alyssa G., Dikeman, Dustin A., Zhang, Jeffrey, Ackerman, Nicole E., Kim, Sophie E., Falgons, Christian, Ortines, Roger V., Liu, Haiyun, Joyce, Daniel P., Alphonse, Martin Prince, Dillen, Carly A., Thompson, John M., Archer, Nathan K., and Miller, Lloyd S.
- Subjects
TUMOR necrosis factors ,INFECTIOUS arthritis ,ANIMAL models in research ,STAPHYLOCOCCUS aureus - Abstract
Orthopedic implant‐associated infection (OIAI) is a major complication that leads to implant failure. In preclinical models of Staphylococcus aureus OIAI, osteomyelitis and septic arthritis, interleukin‐1α (IL‐1α), IL‐1β, and tumor necrosis factor (TNF) are induced, but whether they have interactive or distinctive roles in host defense are unclear. Herein, a S. aureus OIAI model was performed in mice deficient in IL‐1α, IL‐1β, or TNF. Mice deficient in IL‐1β or TNF (to a lesser extent) but not IL‐1α had increased bacterial burden at the site of the OIAI throughout the 28‐day experiment. IL‐1β and TNF had a combined and critical role in host defense as mice deficient in both IL‐1R and TNF (IL‐1R/TNF‐deficient mice) had a 40% mortality rate, which was associated with markedly increased bacterial burden at the site of the OIAI infection. Finally, IL‐1α‐ and IL‐1β‐deficient mice had impaired neutrophil recruitment whereas IL‐1β‐, TNF‐, and IL‐1R/TNF‐deficient mice all had impaired recruitment of both neutrophils and monocytes. Therefore, IL‐1β and TNF contributed to host defense against S. aureus OIAI and neutrophil recruitment was primarily mediated by IL‐1β and monocyte recruitment was mediated by both IL‐1β and TNF. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
37. Clonal Vγ6+Vδ4+ T cells promote IL-17-mediated immunity against Staphylococcus aureus skin infection.
- Author
-
Maverakis, Emanual, Marchitto, Mark C., Dillen, Carly A., Haiyun Liu, Miller, Robert J., Archer, Nathan K., Ortines, Roger V., Alphonse, Martin P., Yu Wang, Pinsker, Bret L., Byrd, Angel S., Brown, Isabelle D., Ravipati, Advaitaa, Zhang, Emily, Cai, Shuting S., Simon, Scott I., Wei Shen, Durum, Scott K., O'Brien, Rebecca L., and Millera, Lloyd S.
- Subjects
STAPHYLOCOCCUS aureus infections ,T cells ,IMMUNITY ,SKIN infections ,INTERLEUKIN-17 ,ANTIBIOTICS ,LYMPH nodes ,NEUTROPHILS - Abstract
T cell cytokines contribute to immunity against Staphylococcus aureus, but the predominant T cell subsets involved are unclear. In an S. aureus skin infection mouse model, we found that the IL- 17 response was mediated by γδ T cells, which trafficked from lymph nodes to the infected skin to induce neutrophil recruitment, proinflammatory cytokines IL-1α, IL-1β, and TNF, and host defense peptides. RNA-seq for TRG and TRD sequences in lymph nodes and skin revealed a single clonotypic expansion of the encoded complementarity-determining region 3 amino acid sequence, which could be generated by canonical nucleotide sequences of TRGV5 or TRGV6 and TRDV4. However, only TRGV6 and TRDV4 but not TRGV5 sequences expanded. Finally, Vγ6+ T cells were a predominant γδ T cell subset that produced IL-17A as well as IL-22, TNF, and IFNγ, indicating a broad and substantial role for clonal Vγ6+Vδ4+ T cells in immunity against S. aureus skin infections. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
38. Mouse model of hematogenous implant-related Staphylococcus aureus biofilm infection reveals therapeutic targets.
- Author
-
Yu Wang, Cheng, Lily I., Helfer, David R., Ashbaugh, Alyssa G., Miller, Robert J., Tzomides, Alexander J., Thompson, John M., Ortines, Roger V., Tsai, Andrew S., Haiyun Liu, Dillen, Carly A., Archer, Nathan K., Cohen, Taylor S., Tkaczyk, Christine, Stover, C. Kendall, Sellman, Bret R., and Miller, Lloyd S.
- Subjects
INFECTION prevention ,MEDICAL equipment design ,ORTHOPEDIC apparatus ,ARTIFICIAL implants ,STAPHYLOCOCCUS aureus genetics - Abstract
Infection is a major complication of implantable medical devices, which provide a scaffold for biofilm formation, thereby reducing susceptibility to antibiotics and complicating treatment. Hematogenous implant-related infections following bacteremia are particularly problematic because they can occur at any time in a previously stable implant. Herein, we developed a model of hematogenous infection in which an orthopedic titanium implant was surgically placed in the legs of mice followed 3 wk later by an i.v. exposure to Staphylococcus aureus. This procedure resulted in a marked propensity for a hematogenous implant-related infection comprised of septic arthritis, osteomyelitis, and biofilm formation on the implants in the surgical legs compared with sham-operated surgical legs without implant placement and with contralateral nonoperated normal legs. Neutralizing human monoclonal antibodies against a-toxin (AT) and clumping factor A (ClfA), especially in combination, inhibited biofilm formation in vitro and the hematogenous implant-related infection in vivo. Our findings suggest that AT and ClfA are pathogenic factors that could be therapeutically targeted against S. aureus hematogenous implant-related infections. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
39. Immunological Methods for Staphylococcus aureus Infection Diagnosis and Prevention.
- Author
-
Archer, Nathan K., Costerton, J. William, Leid, Jeff G., and Shirtliff, Mark E.
- Published
- 2012
- Full Text
- View/download PDF
40. Staphylococcus aureus biofilms.
- Author
-
Archer, Nathan K., Mazaitis, Mark J., Costerton, J. William, Leid, Jeff G., Powers, Mary Elizabeth, and Shirtliff, Mark E.
- Published
- 2011
- Full Text
- View/download PDF
41. Optimization and Validation of an FTIR-based, All-in-one System for Viable MDR Bacteria Detection in Combat-related Wound Infection.
- Author
-
Chen, Ying, Leung, Andrew, Wang, Yulia, and Archer, Nathan K
- Subjects
- *
INSTITUTIONAL review boards , *METHICILLIN-resistant staphylococcus aureus , *MICROBIAL sensitivity tests , *MONONUCLEOSIS , *ANIMAL experimentation - Abstract
Introduction The U.S. Military members experiencing combat-related injuries have a higher chance of developing infections by multidrug-resistant (MDR) bacteria at admission to military hospitals. MDR wound infections result in higher amputation rates and greater risks for subsequent or chronic infections that require readmission or extended stay in the hospital. Currently, there is no FDA-clear, deployable early diagnostic system for suitable field use. We are reporting our efforts to improve a previously developed Rapid Label-free Pathogen Identification (RAPID) system to detect viable MDR bacteria in wound infections and perform antibiotic susceptibility testing (AST). Specifically, we added multiplex and automation capability and significantly simplified the sample preparation process. A functional prototype of the improved system was built, and its performance was validated using a variety of lab-prepared spiked samples and real-world samples. Materials and Methods To access the baseline performance of the improved RAPID system in detecting bacteria presence, we selected 17 isolates, most of them from blood or wound infections, and prepared mono-strain spiked samples at 104 to 106 cfu/mL concentration. These samples were processed and analyzed by the RAPID system. To demonstrate the AST capability of the system, we selected 6 strains against 6 different antibiotics and compared the results from the system with the ones from the gold standard method. To validate the system's performance with real-world samples, we first investigated its performance on 3 swab samples from epicutaneous methicillin-resistant Staphylococcus aureus –exposed mouse model. The AST results from our system were compared with the ones from the gold standard method. All animal experiments were approved by the Johns Hopkins University Animal Care and Use Committee (Protocol No. MO21M378). Then, we obtained swab samples from 7 atopic dermatitis (AD) patients and compared our AST results with the ones from the gold standard method. The human subject protocol was approved by the Johns Hopkins Medicines Institutional Review Boards (Study No. CR00043438/IRB00307926) and by USAMRDC (Proposal Log Number/Study Number 20000251). Results High-quality data were obtained from the spiked samples of all 17 strains. A quantitative analysis model built using these data achieved 94% accuracy in predicting the species ID in 8 unknown samples. The AST results on the spiked samples had shown 100% matching with the gold standard method. Our system successfully detects the presence/absence of viable bacteria in all 3 mouse and 7 AD patient swab samples. Our system shows 100% and 85.7% (6 out of 7) accuracy when compared to the oxacillin susceptibility testing results for the mouse and the AD patient swabs, respectively. Conclusions Our system has achieved excellent performance in detecting viable bacteria presence and in performing AST in a multiplex, automated, and easy-to-operate manner, on both lab-prepared and real samples. Our results have shown a path forward to a rapid (sample-to-answer time ≤3 hours), accurate, sensitive, species-specific, and portable system to detect the presence of MDR combat-related wound infections in the field environment. Our future efforts involve ruggedizing the RAPID system and evaluating performance under relevant environmental conditions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
42. Syndecan-1 Regulates Psoriasiform Dermatitis by Controlling Homeostasis of IL-17-Producing γδ T Cells.
- Author
-
Jaiswal, Anil Kumar, Sadasivam, Mohanraj, Archer, Nathan K., Miller, Robert J., Dillen, Carly A., Ravipati, Advaitaa, Park, Pyong Woo, Chakravarti, Shukti, Miller, Lloyd S., and Hamad, Abdel Rahim A.
- Subjects
- *
SKIN inflammation , *SYNDECANS , *T cells , *HOMEOSTASIS , *APOPTOSIS - Abstract
IL-17 is a potent proinflammatory cytokine that drives pathogenesis of multiple autoimmune diseases, including psoriasis. A major source of pathogenic IL-17 is a subset of γδ T cells (Tγδ17) that acquires the ability to produce IL-17 while developing in the thymus. The mechanisms that regulate homeostasis of Tγδ17 cells and their roles in psoriasis, however, are not fully understood. In this paper, we show that the heparan sulfate proteoglycan syndecan-1 (sdc1) plays a critical role in regulating homeostasis of Tγδ17 cells and modulating psoriasis-like skin inflammation in mice. sdc1 was predominantly expressed by Tγδ17 cells (but not IL-172 Tγδ cells) in the thymus, lymph nodes, and dermis. sdc1 deficiency significantly and selectively increased the frequency and absolute numbers of Tγδ17 cells by mechanisms that included increased proliferation and decreased apoptosis. Adoptive transfer experiments ruled out a significant role of sdc1 expressed on nonhematopoietic cells in halting expansion and proliferation of sdc1-deficient Tγδ17 cells. When subjected to imiquimod-induced psoriasiform dermatitis, Tγδ17 cells in sdc1KO mice displayed heightened responses accompanied by significantly increased skin inflammation than their wild-type counterparts. Furthermore, transferred sdc1-deficient γδ T cells caused more severe psoriasiform dermatitis than their sdc1-sufficient counterparts in TCR-β δ KO hosts. The results uncover a novel role for sdc1 in controlling homeostasis of Tγδ17 cells and moderating host responses to psoriasis-like inflammation. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
43. Commensal microbiome promotes hair follicle regeneration by inducing keratinocyte HIF-1a signaling and glutamine metabolism.
- Author
-
Gaofeng Wang, Sweren, Evan, Andrews, William, Yue Li, Junjun Chen, Yingchao Xue, Wier, Eric, Alphonse, Martin P., Li Luo, Yong Miao, Chen, Ruosi, Dongqiang Zeng, Sam Lee, Ang Li, Dare, Erika, Dongwon Kim, Archer, Nathan K., Reddy, Sashank K., Resar, Linda, and Zhiqi Hu
- Subjects
- *
HAIR follicles , *WOUND healing , *GLUTAMINE synthetase , *GLUTAMINE , *GUT microbiome , *REGENERATION (Biology) , *MOLECULAR biology , *KERATINOCYTES - Abstract
The article presents a study which showed that bacterially induced hypoxia drives increased glutamine metabolism in keratinocytes with attendant enhancement of skin and hair follicle regeneration through comprehensive multiomic analysis and single-cell RNA sequencing in murine skin. Topics discussed include hypoxia and glutamine metabolism in keratinocytes during WIHN, human and mouse keratinocyte isolation, culture, and treatment, and microarray and biofinformatic analysis.
- Published
- 2023
- Full Text
- View/download PDF
44. Keratinocyte-derived defensins activate neutrophil-specific receptors Mrgpra2a/b to prevent skin dysbiosis and bacterial infection.
- Author
-
Dong, Xintong, Limjunyawong, Nathachit, Sypek, Elizabeth I., Wang, Gaofeng, Ortines, Roger V., Youn, Christine, Alphonse, Martin P., Dikeman, Dustin, Wang, Yu, Lay, Mark, Kothari, Ruchita, Vasavda, Chirag, Pundir, Priyanka, Goff, Loyal, Miller, Lloyd S., Lu, Wuyuan, Garza, Luis A., Kim, Brian S., Archer, Nathan K., and Dong, Xinzhong
- Subjects
- *
DEFENSINS , *BACTERIAL diseases , *DYSBIOSIS , *ANTIMICROBIAL peptides , *MACROPHAGE inflammatory proteins - Abstract
Healthy skin maintains a diverse microbiome and a potent immune system to fight off infections. Here, we discovered that the epithelial-cell-derived antimicrobial peptides defensins activated orphan G-protein-coupled receptors (GPCRs) Mrgpra2a/b on neutrophils. This signaling axis was required for effective neutrophil-mediated skin immunity and microbiome homeostasis. We generated mutant mouse lines lacking the entire Defensin (Def) gene cluster in keratinocytes or Mrgpra2a/b. Def and Mrgpra2 mutant animals both exhibited skin dysbiosis, with reduced microbial diversity and expansion of Staphylococcus species. Defensins and Mrgpra2 were critical for combating S. aureus infections and the formation of neutrophil abscesses, a hallmark of antibacterial immunity. Activation of Mrgpra2 by defensin triggered neutrophil release of IL-1β and CXCL2 which are vital for proper amplification and propagation of the antibacterial immune response. This study demonstrated the importance of epithelial-neutrophil signaling via the defensin-Mrgpra2 axis in maintaining healthy skin ecology and promoting antibacterial host defense. [Display omitted] • Orphan GPCRs Mrgpra2a/b are defensin receptors in neutrophils • Defensin-Mrgpra2 signaling preserves skin microbiome diversity • Deletion of defensins or Mrgpra2 impairs neutrophil-mediated bacterial clearance • Activation of Mrgpra2 by defensins triggers neutrophils to release IL-1β and CXCL2 Antimicrobial peptides (AMPs) are vital for host defense. Dong et al. discover that keratinocyte-derived defensins activate G-protein-coupled receptors Mrgpra2a/b on neutrophils. This epithelial-neutrophil signaling axis prevents skin dysbiosis and is required for immunity against bacterial infections. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
45. Dry and liquid formulations of IBT-V02, a novel multi-component toxoid vaccine, are effective against Staphylococcus aureus isolates from low-to-middle income countries.
- Author
-
Wang Y, Mukherjee I, Venkatasubramaniam A, Dikeman D, Orlando N, Zhang J, Ortines R, Mednikov M, Sherchand SP, Kanipakala T, Le T, Shukla S, Ketner M, Adhikari RP, Karauzum H, Aman MJ, and Archer NK
- Subjects
- Animals, Mice, Rabbits, Staphylococcus aureus, Developing Countries, Anti-Bacterial Agents, Bacterial Vaccines, Toxoids, Methicillin-Resistant Staphylococcus aureus, Staphylococcal Infections
- Abstract
Staphylococcus aureus is the leading cause of skin and soft tissue infections (SSTIs) in the U.S. as well as more serious invasive diseases, including bacteremia, sepsis, endocarditis, surgical site infections, osteomyelitis, and pneumonia. These infections are exacerbated by the emergence of antibiotic-resistant clinical isolates such as methicillin-resistant S. aureus (MRSA), highlighting the need for alternatives to antibiotics to treat bacterial infections. We have previously developed a multi-component toxoid vaccine (IBT-V02) in a liquid formulation with efficacy against multiple strains of Staphylococcus aureus prevalent in the industrialized world. However, liquid vaccine formulations are not compatible with the paucity of cold chain storage infrastructure in many low-to-middle income countries (LMICs). Furthermore, whether our IBT-V02 vaccine formulations are protective against S. aureus isolates from LMICs is unknown. To overcome these limitations, we developed lyophilized and spray freeze-dried formulations of IBT-V02 vaccine and demonstrated that both formulations had comparable biophysical attributes as the liquid formulation, including similar levels of toxin neutralizing antibodies and protective efficacy against MRSA infections in murine and rabbit models. To enhance the relevancy of our findings, we then performed a multi-dimensional screen of 83 S. aureus clinical isolates from LMICs (e.g., Democratic Republic of Congo, Palestine, and Cambodia) to rationally down-select strains to test in our in vivo models based on broad expression of IBT-V02 targets (i.e., pore-forming toxins and superantigens). IBT-V02 polyclonal antisera effectively neutralized toxins produced by the S. aureus clinical isolates from LMICs. Notably, the lyophilized IBT-V02 formulation exhibited significant in vivo efficacy in various preclinical infection models against the S. aureus clinical isolates from LMICs, which was comparable to our liquid formulation. Collectively, our findings suggested that lyophilization is an effective alternative to liquid vaccine formulations of our IBT-V02 vaccine against S. aureus infections, which has important implications for protection from S. aureus isolates from LMICs., Competing Interests: Authors IM, AV, RO, MM, SS, TK, RA, HK and MJ were employed by company Integrated Biotherapeutics Inc. Author MK was employed by company Engineered Biopharmaceuticals. Author NA has received previous grant support from Pfizer and Boehringer Ingelheim and has been a paid consultant for Janssen Pharmaceuticals. MJ and RA have stocks in Razi Holdings, Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Wang, Mukherjee, Venkatasubramaniam, Dikeman, Orlando, Zhang, Ortines, Mednikov, Sherchand, Kanipakala, Le, Shukla, Ketner, Adhikari, Karauzum, Aman and Archer.)
- Published
- 2024
- Full Text
- View/download PDF
46. γδ T cell-intrinsic IL-1R promotes survival during Staphylococcus aureus bacteremia.
- Author
-
Wang Y, Ahmadi MZ, Dikeman DA, Youn C, and Archer NK
- Subjects
- Humans, Staphylococcus aureus, Tumor Necrosis Factor-alpha, CD8-Positive T-Lymphocytes, Endothelial Cells, Methicillin-Resistant Staphylococcus aureus physiology, Staphylococcal Infections, Bacteremia prevention & control
- Abstract
Staphylococcus aureus is a leading cause of bacteremia, further complicated by the emergence of antibiotic-resistant strains such as methicillin-resistant S. aureus (MRSA). A better understanding of host defense mechanisms is needed for the development of host-directed therapies as an alternative approach to antibiotics. The levels of IL-1, IL-17, and TNF-α cytokines in circulation have been associated with predictive outcomes in patients with S. aureus bacteremia. However, their causative role in survival and the cell types involved in these responses during bacteremia is not entirely clear. Using a mouse model of S. aureus bacteremia, we demonstrated that IL-17A/F and TNF-α had no significant impact on survival, whereas IL-1R signaling was critical for survival during S. aureus bacteremia. Furthermore, we identified that T cells, but not neutrophils, monocytes/macrophages, or endothelial cells were the crucial cell type for IL-1R-mediated survival against S. aureus bacteremia. Finally, we determined that the expression of IL-1R on γδ T cell, but not CD4
+ or CD8+ T cells was responsible for survival against the S. aureus bacteremia. Taken together, we uncovered a role for IL-1R, but not IL-17A/F and TNF-α in protection against S. aureus bacteremia. Importantly, γδ T cell-intrinsic expression of IL-1R was crucial for survival, but not on other immune cells or endothelial cells. These findings reveal potential cellular and immunological targets for host-directed therapies for improved outcomes against S. aureus bacteremia., Competing Interests: NA has received previous grant support from Pfizer and Boehringer Ingelheim and has been a paid consultant for Janssen Pharmaceuticals. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be constructed as a potential conflict of interest., (Copyright © 2023 Wang, Ahmadi, Dikeman, Youn and Archer.)- Published
- 2023
- Full Text
- View/download PDF
47. Neutrophil-intrinsic TNF receptor signaling orchestrates host defense against Staphylococcus aureus .
- Author
-
Youn C, Pontaza C, Wang Y, Dikeman DA, Joyce DP, Alphonse MP, Wu MJ, Nolan SJ, Anany MA, Ahmadi M, Young J, Tocaj A, Garza LA, Wajant H, Miller LS, and Archer NK
- Subjects
- Humans, Staphylococcus aureus, Receptors, Tumor Necrosis Factor, Type I, Receptors, Tumor Necrosis Factor, Type II, Neutrophils, Staphylococcal Infections drug therapy
- Abstract
Staphylococcus aureus is the leading cause of skin and soft tissue infections and is a major health burden due to the emergence of antibiotic-resistant strains. To address the unmet need of alternative treatments to antibiotics, a better understanding of the protective immune mechanisms against S. aureus skin infection is warranted. Here, we report that tumor necrosis factor (TNF) promoted protection against S. aureus in the skin, which was mediated by bone marrow-derived immune cells. Furthermore, neutrophil-intrinsic TNF receptor (TNFR) signaling directed immunity against S. aureus skin infections. Mechanistically, TNFR1 promoted neutrophil recruitment to the skin, whereas TNFR2 prevented systemic bacterial dissemination and directed neutrophil antimicrobial functions. Treatment with a TNFR2 agonist showed therapeutic efficacy against S. aureus and Pseudomonas aeruginosa skin infections, which involved increased neutrophil extracellular trap formation. Our findings revealed nonredundant roles for TNFR1 and TNFR2 in neutrophils for immunity against S. aureus and can be therapeutically targeted for protection against bacterial skin infections.
- Published
- 2023
- Full Text
- View/download PDF
48. Commensal microbiome promotes hair follicle regeneration by inducing keratinocyte HIF-1α signaling and glutamine metabolism.
- Author
-
Wang G, Sweren E, Andrews W, Li Y, Chen J, Xue Y, Wier E, Alphonse MP, Luo L, Miao Y, Chen R, Zeng D, Lee S, Li A, Dare E, Kim D, Archer NK, Reddy SK, Resar L, Hu Z, Grice EA, Kane MA, and Garza LA
- Subjects
- Animals, Humans, Mice, Keratinocytes, Regeneration, Skin metabolism, Wound Healing, Microbiota, Glutamine metabolism, Hair Follicle
- Abstract
Tissue injury induces metabolic changes in stem cells, which likely modulate regeneration. Using a model of organ regeneration called wound-induced hair follicle neogenesis (WIHN), we identified skin-resident bacteria as key modulators of keratinocyte metabolism, demonstrating a positive correlation between bacterial load, glutamine metabolism, and regeneration. Specifically, through comprehensive multiomic analysis and single-cell RNA sequencing in murine skin, we show that bacterially induced hypoxia drives increased glutamine metabolism in keratinocytes with attendant enhancement of skin and hair follicle regeneration. In human skin wounds, topical broad-spectrum antibiotics inhibit glutamine production and are partially responsible for reduced healing. These findings reveal a conserved and coherent physiologic context in which bacterially induced metabolic changes improve the tolerance of stem cells to damage and enhance regenerative capacity. This unexpected proregenerative modulation of metabolism by the skin microbiome in both mice and humans suggests important methods for enhancing regeneration after injury.
- Published
- 2023
- Full Text
- View/download PDF
49. Staphylococcus aureus adaptive evolution: Recent insights on how immune evasion, immunometabolic subversion and host genetics impact vaccine development.
- Author
-
Wong Fok Lung T, Chan LC, Prince A, Yeaman MR, Archer NK, Aman MJ, and Proctor RA
- Subjects
- Humans, Staphylococcus aureus genetics, Immune Evasion, Prospective Studies, Staphylococcal Infections microbiology, Vaccines
- Abstract
Despite meritorious attempts, a S. aureus vaccine that prevents infection or mitigates severity has not yet achieved efficacy endpoints in prospective, randomized clinical trials. This experience underscores the complexity of host- S. aureus interactions, which appear to be greater than many other bacterial pathogens against which successful vaccines have been developed. It is increasingly evident that S. aureus employs strategic countermeasures to evade or exploit human immune responses. From entering host cells to persist in stealthy intracellular reservoirs, to sensing the environmental milieu and leveraging bacterial or host metabolic products to reprogram host immune responses, S. aureus poses considerable challenges for the development of effective vaccines. The fact that this pathogen causes distinct types of infections and can undergo transient genetic, transcriptional or metabolic adaptations in vivo that do not occur in vitro compounds challenges in vaccine development. Notably, the metabolic versatility of both bacterial and host immune cells as they compete for available substrates within specific tissues inevitably impacts the variable repertoire of gene products that may or may not be vaccine antigens. In this respect, S. aureus has chameleon phenotypes that have alluded vaccine strategies thus far. Nonetheless, a number of recent studies have also revealed important new insights into pathogenesis vulnerabilities of S. aureus . A more detailed understanding of host protective immune defenses versus S. aureus adaptive immune evasion mechanisms may offer breakthroughs in the development of effective vaccines, but at present this goal remains a very high bar. Coupled with the recent advances in human genetics and epigenetics, newer vaccine technologies may enable such a goal. If so, future vaccines that protect against or mitigate the severity of S. aureus infections are likely to emerge at the intersection of precision and personalized medicine. For now, the development of S. aureus vaccines or alternative therapies that reduce mortality and morbidity must continue to be pursued., Competing Interests: RP is a consultant for IBT which is producing a multi-valent antitoxin vaccine and is a member of the review board for the University of Rochester which is involved in an anti-Gmd vaccine effort. MY is a consultant for Genentech-Roche, Alexion/AstraZeneca and Horizon Pharmaceuticals, which are involved in discovery and development of immunotherapeutic agents and strategies. NA is a consultant for Janssen Pharmaceuticals and has received previous grant support from Pfizer and Boehringer Ingelheim, which are involved in discovery and development of immunotherapeutic agents and strategies. MA has stocks in Integrated Biotherapeutics, a company engaged in development of vaccines and antibody therapies for bacterial infections. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Wong Fok Lung, Chan, Prince, Yeaman, Archer, Aman and Proctor.)
- Published
- 2022
- Full Text
- View/download PDF
50. The Novel Oxazolidinone TBI-223 Is Effective in Three Preclinical Mouse Models of Methicillin-Resistant Staphylococcus aureus Infection.
- Author
-
Gordon O, Dikeman DA, Ortines RV, Wang Y, Youn C, Mumtaz M, Orlando N, Zhang J, Patel AM, Gough E, Kaushik A, Nuermberger EL, Upton AM, Fotouhi N, Miller LS, and Archer NK
- Subjects
- Animals, Mice, Acetamides pharmacology, Acetamides therapeutic use, Anti-Bacterial Agents adverse effects, Linezolid adverse effects, Microbial Sensitivity Tests, Staphylococcus aureus, Bacteremia drug therapy, Methicillin-Resistant Staphylococcus aureus, Oxazolidinones adverse effects, Staphylococcal Infections drug therapy, Staphylococcal Infections microbiology
- Abstract
Staphylococcus aureus is an important cause of various infections in humans, including bacteremia, skin and soft tissue infections, and infections associated with implanted medical devices. The emergence of hospital- and community-acquired methicillin-resistant Staphylococcus aureus (MRSA) underscores the urgent and unmet need to develop novel, safe, and effective antibiotics against these multidrug-resistant clinical isolates. Oxazolidinone antibiotics such as linezolid have excellent oral bioavailability and provide coverage against MRSA infections. However, their widespread and long-term use is often limited by adverse effects, especially myelosuppression. TBI-223 is a novel oxazolidinone with potentially reduced myelosuppression, compared to linezolid, but its efficacy against MRSA infections is unknown. Therefore, the preclinical efficacy of TBI-223 (80 and 160 mg/kg twice daily) was compared with that of linezolid (40 and 80 mg/kg twice daily) and sham treatment in mouse models of MRSA bacteremia, skin wound infection, and orthopedic-implant-associated infection. The dosage was selected based on mouse pharmacokinetic analysis of both linezolid and TBI-223, as well as measurement of the MICs. In all three models, TBI-223 and linezolid had comparable dose-dependent efficacies in reducing bacterial burden and disease severity, compared with sham-treated control mice. Taken together, these findings indicate that TBI-223 represents a novel oxazolidinone antibiotic that may provide an additional option against MRSA infections. Future studies in larger animal models and clinical trials are warranted to translate these findings to humans. IMPORTANCE Staphylococcus aureus is the predominant cause of bloodstream, skin, and bone infections in humans. Resistance to commonly used antibiotics is a growing concern, making it more difficult to treat staphylococcal infections. Use of the oxazolidinone antibiotic linezolid against resistant strains is hindered by high rates of adverse reactions during prolonged therapy. Here, a new oxazolidinone named TBI-223 was tested against S. aureus in three mouse models of infection, i.e., bloodstream infection, skin infection, and bone infection. We found that TBI-223 was as effective as linezolid in these three models. Previous data suggest that TBI-223 has a better safety profile than linezolid. Taken together, these findings indicate that this new agent may provide an additional option against MRSA infections. Future studies in larger animal models and clinical trials are warranted to translate these findings to humans.
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.