1
|
Wen J, Xu X, Su Y, Qi X, Fu Q, Chen M, Wang X. Impact of doxycycline on mycoplasma pneumonia treatment and cancer prognosis in pediatric leukemia patients post-chemotherapy: a target trial emulation. Am J Cancer Res 2025; 15:1790-1805. [PMID: 40371165 PMCID: PMC12070108 DOI: 10.62347/kgbq9641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/19/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Acute leukemia is the most common malignancy in children. While chemotherapy is effective, it significantly compromises immune function, leading to a high incidence of infectious complications, such as secondary pneumonia, particularly Mycoplasma pneumonia (MP). The treatment of infections in pediatric leukemia patients faces challenges such as antibiotic resistance and drug interactions during chemotherapy. OBJECTIVE This study aims to evaluate the therapeutic efficacy and safety of doxycycline in treating MP in pediatric leukemia patients post-chemotherapy, as well as it impact on chemotherapy continuity. METHODS This study employed a target trial emulation design using retrospective data from pediatric leukemia patients diagnosed with MP. Patients aged 12-17 years with confirmed leukemia and clinical evidence of pneumonia following chemotherapy were included. Doxycycline was compared to azithromycin and other empirical treatments. Follow-up assessments at 3 days, 5 days, 30 days, and 180 days evaluated fever resolution, radiological improvement, additional interventions, and adverse events. Statistical analyses included Kaplan-Meier survival analysis and Cox proportional hazards models. RESULTS In Trial 2, doxycycline demonstrated a significantly higher treatment success rate than other empirical treatments (87.72% vs. 73.13%, P = 0.013) and was associated with faster fever resolution (P = 0.048) and shorter time to chest X-ray improvement (P = 0.048). The 30-day survival rate was significantly higher in the Doxycycline group compared to other empirical treatments (100% vs. 91.04%, P = 0.019). Fewer patients require additional interventions such as ICU admission (P = 0.019). Furthermore, patients in the Doxycycline group had a significantly higher likelihood of completing chemotherapy without delays (84.21% vs. 59.70%, P = 0.01). In Trial 1, no significant differences were observed in treatment success rate, fever resolution time, hospitalization duration, or chemotherapy tolerance between Doxycycline and Azithromycin (P > 0.05). CONCLUSION Doxycycline, as a broad-spectrum antibiotic, demonstrates efficacy comparable to azithromycin in treating MP with advantages in reducing chemotherapy-related delays, hospitalization duration, and the need for additional interventions. It enhances chemotherapy tolerance and continuity. Doxycycline may serve as an economical and effective alternative for managing post-chemotherapy infections in pediatric leukemia patients, especially in cases of antibiotic resistance or intolerance.
Collapse
Affiliation(s)
- Jun Wen
- Emergency Department, Xi’an Children’s HospitalXi’an 710000, Shaanxi, China
| | - Xiaoyan Xu
- Emergency Department, Xi’an Children’s HospitalXi’an 710000, Shaanxi, China
| | - Yufei Su
- Emergency Department, Xi’an Children’s HospitalXi’an 710000, Shaanxi, China
| | - Xuehao Qi
- Emergency Department, Xi’an Children’s HospitalXi’an 710000, Shaanxi, China
| | - Qiang Fu
- Hematology Department, Xi’an Children’s HospitalXi’an 710000, Shaanxi, China
| | - Mingjiang Chen
- Information Department, Xi’an Children’s HospitalXi’an 710000, Shaanxi, China
| | - Xue Wang
- Emergency Department, Xi’an Children’s HospitalXi’an 710000, Shaanxi, China
| |
Collapse
|
2
|
Tian T, Xue Z, Sun X, Ding L, Zhao R, Wang Z, Wu J, Li X, Li Y, Sun L. Qingfei Yin alleviates Streptococcus pneumoniae pneumonia by promoting complete autophagy to suppress necroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156280. [PMID: 39637472 DOI: 10.1016/j.phymed.2024.156280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/21/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Bacterial pneumonia is most prevalent among all pneumonia types, with Streptococcus pneumoniae being the main pathogen. Qingfei Yin (QFY) is a traditional Chinese medicine formula used in the clinical treatment of bacterial pneumonia. Previous studies have confirmed the multi-target and -effect characteristics of QFY in treating S. pneumoniae pneumonia. PURPOSE The purpose of this study was to explore the mechanism underlying QFY in treating pneumonia produced by S. pneumoniae. METHODS First, an in vivo model of S. pneumoniae-induced pneumonia was established in mice and evaluated the efficacy of QFY by hematoxylin-eosin (HE) staining and measuring cytokine levels in bronchoalveolar lavage fluid. Next, single-cell transcriptomics was used to identify the targeted cell subtypes, signaling pathways, and biological processes affected by QFY. Finally, the findings were validated using a pneumolysin (PLY) -induced mouse lung epithelial cells (TC-1) model in vitro using western blot analysis, immunofluorescence (IF), acridine orange (AO) staining, and propidium iodide (PI) staining. RESULTS QFY was shown to alleviate lung inflammation and reduce the TNF-α and IL-6 levels in bronchoalveolar lavage fluid in vivo. A total of 113,353 cells were classified using single-cell transcriptomics and 12 major cell types were identified. By single-cell transcriptomics, QFY was confirmed to primarily target lung epithelial cells. Differentially expressed genes were shown to be enriched in autophagy and necroptosis signaling pathways, and the key differentially expressed gene, Sequestosome 1 (p62/SQSTM1), was identified. PLY was shown to induce RIPK1-dependent necroptosis and incomplete autophagy in TC-1 cells. QFY was shown to promote complete autophagy by downregulating the expression of p62, thereby reducing phosphorylation of RIPK1 and MLKL, and alleviating necroptosis in S. pneumoniae-induced lung epithelial cell death. CONCLUSION This study demonstrated that QFY can effectively alleviate S. pneumoniae pneumonia. The mechanism of action may be that QFY promotes complete autophagy by downregulating p62 expression, thereby alleviating necroptosis of S. pneumoniae-induced lung epithelial cells and reducing lung injury. It provides a scientific basis for clinical prevention and treatment of S. pneumoniae pneumonia.
Collapse
Affiliation(s)
- Tong Tian
- Traditional Chinese Medicine College, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Zhilong Xue
- Traditional Chinese Medicine College, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Xiaozhou Sun
- Pediatric Respiratory Medicine Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, PR China
| | - Lizhong Ding
- Pediatric Respiratory Medicine Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, PR China
| | - Renshuang Zhao
- Medical College, Yanbian University, Yanji 133002, PR China
| | - Zhongtian Wang
- Traditional Chinese Medicine College, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Jiaqi Wu
- Traditional Chinese Medicine College, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Xiao Li
- Medical College, Yanbian University, Yanji 133002, PR China; Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun 130117, PR China.
| | - Yiquan Li
- Medical College, Yanbian University, Yanji 133002, PR China; Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun 130117, PR China.
| | - Liping Sun
- Pediatric Respiratory Medicine Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, PR China.
| |
Collapse
|
3
|
Rahayu YC, Setiawatie EM, Rahayu RP, Siswandono S, Indrawati R, Budi HS, Notobroto HB, Darojah RAA. Anti-Osteoclastogenesis Potential of Cocoa Pod Husk (Theobroma cacao L.) Extract: In Silico and In Vivo Study. Braz Dent J 2024; 35:e246015. [PMID: 39630805 DOI: 10.1590/0103-6440202406015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/06/2024] [Indexed: 12/07/2024] Open
Abstract
Periodontitis is a common chronic inflammatory disease characterized by alveolar bone loss. The high polyphenol content in cocoa pod husk (Theobroma cacao L) has the potential to influence bone metabolism and contribute to the inhibition of bone resorption. The aim of this study was to analyze the anti-osteoclastogenesis potential of cocoa pod husk (Theobroma cacao L.) in both in silico and in vivo study. An analysis of the anti-osteoclastogenesis potential of T. cacao bioactive compounds was conducted using molecular docking simulations. Thirty male Wistar rats (Rattus novergicus) were randomly assigned to control negative groups (placebo gel), control positive groups (2% doxycycline gel), and treatment groups (10% cocoa pod husk (CPH) extract gel), with measurements taken on days 7 and 14. Wistar rats were induced with 0.05 ml of P. gingivalis at a concentration of 2x109 CFU/ml intrasulcularly in the maxillary molar to achieved in periodontitis. The number of osteoclasts was observed by hematoxylin and eosin staining, the level of TNF-α was assessed by enzyme-linked immunosorbent assay, and the expression of RANKL was evaluated by immunohistochemical staining. Data were analyzed using One-way ANOVA to examine the differences between the groups. The in silico study showed that the catechin, epicatechin, quercetin, and procyanidin B2 had a strong binding affinity for TNF-α and RANKL. Administration of 10% CPH reduced the number of osteoclasts (p<0.05), TNF-α level on days 7 and 14 (p<0.05), and RANKL expression on day 7 (p<0.05) in experimental rats with periodontitis. Administering 10% CPH inhibited osteoclastogenesis in the experimental periodontitis rats.
Collapse
Affiliation(s)
- Yani Corvianindya Rahayu
- Doctoral Study Program in Dental Science, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Oral Biology, Faculty of Dentistry, Universitas Jember, Indonesia
| | | | - Retno Pudji Rahayu
- Department of Oral Pathology and Maxillofacial, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - S Siswandono
- Department of Pharmacology, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Retno Indrawati
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Hendrik Setia Budi
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Hari Basuki Notobroto
- Department of Epidemiology, Biostatistics and Population Studies, and Health Promotion, Faculty of Public Health, Universitas Airlangga, Surabaya, Indonesia
| | | |
Collapse
|
4
|
Gross JL, Basu R, Bradfield CJ, Sun J, John SP, Das S, Dekker JP, Weiss DS, Fraser IDC. Bactericidal antibiotic treatment induces damaging inflammation via TLR9 sensing of bacterial DNA. Nat Commun 2024; 15:10359. [PMID: 39609397 PMCID: PMC11605096 DOI: 10.1038/s41467-024-54497-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
The immunologic consequences of using bactericidal versus bacteriostatic antibiotic treatments are unclear. We observed a bacteriostatic (growth halting) treatment was more protective than a bactericidal (bacteria killing) treatment in a murine peritonitis model. To understand this unexpected difference, we compared macrophage responses to bactericidal treated bacteria or bacteriostatic treated bacteria. We found that Gram-negative bacteria treated with bactericidal drugs induced more proinflammatory cytokines than those treated with bacteriostatic agents. Bacterial DNA - released only by bactericidal treatments - exacerbated inflammatory signaling through TLR9. Without TLR9 signaling, the in vivo efficacy of bactericidal drug treatment was rescued. This demonstrates that antibiotics can act in important ways distinct from bacterial inhibition: like causing treatment failure by releasing DNA that induces excessive inflammation. These data establish a novel link between how an antibiotic affects bacterial physiology and subsequent immune system engagement, which may be relevant for optimizing treatments to simultaneously clear bacteria and modulate inflammation.
Collapse
Affiliation(s)
- Julia L Gross
- Emory University/NIAID Graduate Partnership Program, Bethesda, MD, USA
| | - Rahul Basu
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, Bethesda, MD, USA
| | - Clinton J Bradfield
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, Bethesda, MD, USA
| | - Jing Sun
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, Bethesda, MD, USA
| | - Sinu P John
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, Bethesda, MD, USA
| | - Sanchita Das
- Department of Laboratory Medicine, NIH Clinical Center, NIH, Bethesda, MD, USA
| | - John P Dekker
- Department of Laboratory Medicine, NIH Clinical Center, NIH, Bethesda, MD, USA
- Bacterial Pathogenesis and Antimicrobial Resistance Unit, Laboratory of Clinical Immunology and Microbiology, NIAID, Bethesda, MD, USA
| | - David S Weiss
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA.
- Emory Antibiotic Resistance Center, Atlanta, GA, USA.
| | - Iain D C Fraser
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, Bethesda, MD, USA.
| |
Collapse
|
5
|
Palillo MB, Carrasco SE, Mishkin N, Palillo JA, Lynch DB, Lawton S, Aydin M, Mourino A, Lipman NS, Ricart Arbona RJ. Assessment of Antimicrobial Therapy in Eradicating Chlamydia muridarum in Research Mice: Immune Status and its Impact on Outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.600682. [PMID: 38979332 PMCID: PMC11230361 DOI: 10.1101/2024.06.28.600682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Chlamydia muridarum (Cm) is a moderately prevalent, gram-negative, intracellular bacterium that affects laboratory mice, causing subclinical to severe disease, depending on the host's immune status. The effectiveness of various antibiotic regimens aimed at eradicating Cm in both immunodeficient and immunocompetent laboratory mice was evaluated. NSG mice were cohoused with Cm-shedding BALB/cJ mice for 14 days to simulate natural exposure. Four groups of 8 infected NSG mice were treated for 7 days with either 0.08% sulfamethoxazole and 0.016% trimethoprim (TMS) in water, 0.0625% doxycycline in feed, 0.124%/0.025% TMS in feed, or 0.12% amoxicillin in feed. A control group was provided standard water and feed. The impact of treatment on gastrointestinal microbiota (GM) was performed using next-generation shotgun sequencing on the last day of treatment. TMS and Amoxicillin had negligible effects on GM, while doxycycline had the largest effect. All antibiotic treated NSG mice exhibited clinical disease, including dehydration, hunched posture, >20% weight loss, and dyspnea, leading to euthanasia 21-40 days post-treatment (32.6 ± 4.2 days; mean ± SD). Untreated controls were euthanized 14-33 days post-exposure (23.75 ± 5.9 days). All mice were fecal PCR positive for Cm at euthanasia. Histological evaluation revealed multifocal histiocytic and neutrophilic bronchointerstitial pneumonia and/or bronchiolitis featuring prominent intralesional chlamydial inclusion bodies in all mice. Subsequently, groups of 8 C57BL/6J, BALB/cJ, NOD.SCID, and NSG mice infected with Cm were treated with 0.124%/0.025% TMS in feed for 7 (BALB/cJ and C57BL/6J) or 21 days (NSG and NOD.SCID). All immunocompetent and NOD.SCID mice were negative for Cm by PCR 14 days post-treatment, remained clinically normal and had no evidence of Cm infection at necropsy, all NSG mice remained Cm positive and were euthanized. While these findings highlight the difficulties in eradicating Cm from highly immunodeficient mice, eradication of Cm from immunocompetent or moderately immunocompromised mice with antibiotics is feasible.
Collapse
|
6
|
Kim DH, Shin H, Stybayeva G, Hwang SH. A comparison of doxycycline and conventional treatments of refractory chronic rhinosinusitis with nasal polyps: a systematic review and meta-analysis. Eur Arch Otorhinolaryngol 2024; 281:3949-3957. [PMID: 38512383 DOI: 10.1007/s00405-024-08563-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/15/2024] [Indexed: 03/23/2024]
Abstract
PURPOSE To compare the effects of doxycycline (DOX) and conventional management in patients with refractory chronic rhinosinusitis and nasal polyps (CRSwNP). METHODS Six databases were searched to September 2023. We retrieved studies that compared improvements in refractory chronic sinusitis-related symptoms between DOX-treated and control groups. RESULTS DOX significantly reduced the Lund-Kennedy (LK) score [- 0.3670 (range - 0.6173; - 0.1166); I2 = 92.8%], the nasal polyposis score [- 0.9484 (- 1.2287; - 0.6680); I2 = 92.5%], the patient-reported Sinonasal Outcome Test (SNOT) score [- 0.3141 (- 0.4622; - 0.1660); I2 = 91.2%], and the nasal obstruction score [- 0.1813 (- 0.3382; - 0.0243); I2 = 86.2%]. On subgroup analyses by the measurement timepoints, the extent of nasal polyposis was significantly lower in the DOX group during treatment, at the end of treatment, and 4 and 8 weeks later. The LK scores also indicated improvements during treatment and at the end of treatment. The SNOT score tended to decrease with time in the treatment group. Nasal obstruction symptoms improved during treatment and 4 weeks later. CONCLUSION DOX enhances the postoperative endoscopic outcomes of refractory CRSwNP patients by reducing recurrent polyposis and inflammation.
Collapse
Affiliation(s)
- Do Hyun Kim
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Hyesoo Shin
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Se Hwan Hwang
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Bucheon St. Mary's Hospital, The Catholic University of Korea, 327 Sosa-ro, Bucheon-si, Gyeonggi-do, Seoul, 14647, Korea.
| |
Collapse
|
7
|
Nikolajevic N, Nikolajevic M, Pantic I, Korica B, Kotseva M, Alempijevic T, Jevtic D, Madrid CI, Dumic I. Drug-Induced Liver Injury Due to Doxycycline: A Case Report and Review of Literature. Cureus 2024; 16:e59687. [PMID: 38836151 PMCID: PMC11150051 DOI: 10.7759/cureus.59687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
Antibiotics are among the most common causes of drug-induced liver injury worldwide. Amoxicillin/clavulanic acid and nitrofurantoin are the most common culprits while tetracyclines are a rare cause of liver injury. Among tetracyclines, minocycline has been reported more frequently than doxycycline, which is an extremely rare cause of drug-induced liver injury. We present a healthy 28-year-old male patient from rural United States who was taking doxycycline for Lyme disease. After five days of therapy, he developed nausea, vomiting, fatigue, and significant transaminitis consistent with a hepatocellular pattern of liver injury. After a thorough workup which ruled out other causes such as infection, autoimmune diseases, liver malignancy, and vascular, structural, and metabolic disorders, his liver injury was attributed to doxycycline. We reached the diagnosis also by demonstrating a consistent temporal association between doxycycline intake and liver injury and the patient recovered completely with the cessation of doxycycline. Recognition of doxycycline as a cause of drug-induced liver injury should be considered in patients utilizing this antibiotic. Doxycycline, unlike minocycline, has a short latency period. Early recognition and discontinuation of doxycycline in our patient resulted in the complete resolution of symptoms and transaminitis preventing further morbidity and mortality.
Collapse
Affiliation(s)
- Nikola Nikolajevic
- Internal Medicine, University of Belgrade, Faculty of Medicine, Belgrade, SRB
| | - Milan Nikolajevic
- Internal Medicine, University of Belgrade, Faculty of Medicine, Belgrade, SRB
| | - Ivana Pantic
- Gastroenterology and Hepatology, Clinic for Gastroenterology, University Clinical Center of Serbia, Belgrade, SRB
| | - Bojan Korica
- Gastroenterology and Hepatology, Clinic for Gastroenterology, University Clinical Center of Serbia, Belgrade, SRB
| | | | - Tamara Alempijevic
- Gastroenterology and Hepatology, Clinic for Gastroenterology, University Clinical Center of Serbia, Belgrade, SRB
| | - Dorde Jevtic
- Internal Medicine, NYC Health + Hospitals/Elmhurst, Queens, USA
| | | | - Igor Dumic
- Hospital Medicine, Mayo Clinic Health System, Eau Claire, USA
| |
Collapse
|
8
|
Ham SD, Abraham MN, Deutschman CS, Taylor MD. Single-cell RNA sequencing reveals Immune Education promotes T cell survival in mice subjected to the cecal ligation and puncture sepsis model. Front Immunol 2024; 15:1366955. [PMID: 38562928 PMCID: PMC10982361 DOI: 10.3389/fimmu.2024.1366955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
Background Individual T cell responses vary significantly based on the microenvironment present at the time of immune response and on prior induced T cell memory. While the cecal ligation and puncture (CLP) model is the most commonly used murine sepsis model, the contribution of diverse T cell responses has not been explored. We defined T cell subset responses to CLP using single-cell RNA sequencing and examined the effects of prior induced T cell memory (Immune Education) on these responses. We hypothesized that Immune Education prior to CLP would alter T cell responses at the single cell level at a single, early post-CLP time point. Methods Splenic T cells were isolated from C57BL/6 mice. Four cohorts were studied: Control, Immune-Educated, CLP, and Immune-Educated CLP. At age 8 weeks, Immune-Educated and Immune-Educated CLP mice received anti-CD3ϵ antibody; Control and CLP mice were administered an isotype control. CLP (two punctures with a 22-gauge needle) was performed at 12-13 weeks of life. Mice were sacrificed at baseline or 24-hours post-CLP. Unsupervised clustering of the transcriptome library identified six distinct T cell subsets: quiescent naïve CD4+, primed naïve CD4+, memory CD4+, naïve CD8+, activated CD8+, and CD8+ cytotoxic T cell subsets. T cell subset specific gene set enrichment analysis and Hurdle analysis for differentially expressed genes (DEGs) were performed. Results T cell responses to CLP were not uniform - subsets of activated and suppressed T cells were identified. Immune Education augmented specific T cell subsets and led to genomic signatures favoring T cell survival in unoperated and CLP mice. Additionally, the combination of Immune Education and CLP effected the expression of genes related to T cell activity in ways that differed from CLP alone. Validating our finding that IL7R pathway markers were upregulated in Immune-Educated CLP mice, we found that Immune Education increased T cell surface IL7R expression in post-CLP mice. Conclusion Immune Education enhanced the expression of genes associated with T cell survival in unoperated and CLP mice. Induction of memory T cell compartments via Immune Education combined with CLP may increase the model's concordance to human sepsis.
Collapse
Affiliation(s)
- Steven D. Ham
- The Division of Critical Care Medicine, Department of Pediatrics, Cohen Children’s Medical Center/Northwell Health, New Hyde Park, NY, United States
- Sepsis Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Mabel N. Abraham
- The Division of Critical Care Medicine, Department of Pediatrics, Cohen Children’s Medical Center/Northwell Health, New Hyde Park, NY, United States
- Sepsis Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Clifford S. Deutschman
- The Division of Critical Care Medicine, Department of Pediatrics, Cohen Children’s Medical Center/Northwell Health, New Hyde Park, NY, United States
- Sepsis Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Matthew D. Taylor
- The Division of Critical Care Medicine, Department of Pediatrics, Cohen Children’s Medical Center/Northwell Health, New Hyde Park, NY, United States
- Sepsis Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| |
Collapse
|
9
|
Alblowi JA, Farid ZS, Attia MS. Comparative Study of Azithromycin Versus Doxycycline Effect on the Resistin Level in Periodontitis Patients With Type 2 Diabetes: A Randomized Controlled Clinical Trial. Cureus 2024; 16:e54849. [PMID: 38533160 PMCID: PMC10964125 DOI: 10.7759/cureus.54849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2024] [Indexed: 03/28/2024] Open
Abstract
AIM The present study aimed to determine if azithromycin (AZM) and doxycycline therapy, as an adjunct to scaling and root planning (SRP), modulate host response and improve clinical outcomes in periodontitis patients with type 2 diabetes mellitus (T2DM). PATIENTS AND METHODS Forty-five periodontal sites in 15 periodontitis patients with T2DM received nonsurgical periodontal therapy (NSPT). In Group I, patients were placebo (not receiving any medication), Group II patients received systemic AZM therapy (AZM 250 mg/day for five days), and Group III patients received doxycycline (20 mg twice per day for three months. The resistin level was collected and measured by enzyme-linked immunosorbent assay (ELISA). Gingival index (GI), probing depth (PD), and clinical attachment level (CAL) were recorded at baseline, one-month, and three-month intervals. RESULTS All groups showed improvement in clinical parameters and resistin levels throughout the study. The mean resistin level at three months was the highest in Group I and the lowest in Group III. Patients in Group II showed a larger decrease in mean PD than those in Group I and III. Group III had the highest gain in mean CAL, with an increase of 1.78 mm in attachment. CONCLUSION Resistin might be a useful indicator of current disease status. In addition, benefits from adjunctive systemic use of AZM and doxycycline have been administered with non-surgical periodontal therapy.
Collapse
Affiliation(s)
- Jazia A Alblowi
- Department of Periodontology, Faculty of Dentistry, King Abdulaziz University, Jeddah, SAU
| | - Zienab S Farid
- Department of Oral Medicine, Periodontology, Diagnosis and Radiology, Faculty of Dental Medicine, Al-Azhar University (Girls Branch), Cairo, EGY
| | - Mai S Attia
- Department of Oral Medicine, Periodontology, Diagnosis and Radiology, Faculty of Dental Medicine, Al-Azhar University (Girls Branch), Cairo, EGY
- Department of Oral Medicine, Periodontology, Diagnosis and Radiology, Faculty of Dental Medicine, Misr International University, Cairo, EGY
| |
Collapse
|
10
|
Morsy SAA, Fathelbab MH, El-Sayed NS, El-Habashy SE, Aly RG, Harby SA. Doxycycline-Loaded Calcium Phosphate Nanoparticles with a Pectin Coat Can Ameliorate Lipopolysaccharide-Induced Neuroinflammation Via Enhancing AMPK. J Neuroimmune Pharmacol 2024; 19:2. [PMID: 38236457 PMCID: PMC10796490 DOI: 10.1007/s11481-024-10099-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024]
Abstract
Neuroinflammation occurs in response to different injurious triggers to limit their hazardous effects. However, failure to stop this process can end in multiple neurological diseases. Doxycycline (DX) is a tetracycline, with potential antioxidant and anti-inflammatory properties. The current study tested the effects of free DX, DX-loaded calcium phosphate (DX@CaP), and pectin-coated DX@CaP (Pec/DX@CaP) nanoparticles on the lipopolysaccharide (LPS)-induced neuroinflammation in mice and to identify the role of adenosine monophosphate-activated protein kinase (AMPK) in this effect. The present study was conducted on 48 mice, divided into 6 groups, eight mice each. Group 1 (normal control), Group 2 (blank nanoparticles-treated), Group 3 (LPS (untreated)), Groups 4, 5, and 6 received LPS, then Group 4 received free DX, Group 5 received DX-loaded calcium phosphate nanoparticles (DX@CaP), and Group 6 received DX-loaded calcium phosphate nanoparticles with a pectin coat (Pec/DX@CaP). At the end of the experimentation period, behavioral tests were carried out. Then, mice were sacrificed, and brain tissue was extracted and used for histological examination, and assessment of interleukin-6 positive cells in different brain areas, in addition to biochemical measurement of SOD activity, TLR-4, AMPK and Nrf2. LPS can induce prominent neuroinflammation. Treatment with (Pec/DX@CaP) can reverse most behavioral, histopathological, and biochemical changes caused by LPS. The findings of the current study suggest that (Pec/DX@CaP) exerts a significant reverse of LPS-induced neuroinflammation by enhancing SOD activity, AMPK, and Nrf2 expression, in addition to suppression of TLR-4.
Collapse
Affiliation(s)
| | - Mona Hassan Fathelbab
- Medical Biochemistry Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Norhan S El-Sayed
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Salma E El-Habashy
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Rania G Aly
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Sahar A Harby
- Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
11
|
Sordi R, Bojko L, Oliveira FRMB, Rosales TO, Souza CF, Moreno LW, Ferreira Alves G, Vellosa JCR, Fernandes D, Gomes JR. Doxycycline reduces liver and kidney injuries in a rat hemorrhagic shock model. Intensive Care Med Exp 2024; 12:2. [PMID: 38194181 PMCID: PMC10776514 DOI: 10.1186/s40635-023-00586-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/18/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Hemorrhagic shock (HS), which causes insufficient tissue perfusion, can result in multiple organ failure (MOF) and death. This study aimed to evaluate whether doxycycline (DOX) protects cardiovascular, kidney, and liver tissue from damage in a rat model of HS. Immediately before the resuscitation, DOX (10 mg/kg; i.v.) was administered, and its protective effects were assessed 24 h later. Mean arterial pressure, renal blood flow, heart rate, vasoactive drug response, and blood markers such as urea, creatinine, AST, ALT, CPK, CPR, and NOx levels were determined. RESULTS We showed that DOX has a significant effect on renal blood flow and on urea, creatinine, AST, ALT, CPK, and NOx. Morphologically, DOX reduced the inflammatory process in the liver tissue. CONCLUSIONS We conclude that DOX protects the liver and kidney against injury and dysfunction in a HS model and could be a strategy to reduce organ damage associated with ischemia-and-reperfusion injury.
Collapse
Affiliation(s)
- Regina Sordi
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Department of Structural Biology, Molecular and Genetics, Post Graduation Program in Biomedical Science, Universidade Estadual de Ponta Grossa, Avenida Carlos Cavalcanti, 4748, Ponta Grossa, PR, 84030-900, Brazil
| | - Luana Bojko
- Department of Structural Biology, Molecular and Genetics, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Filipe R M B Oliveira
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Thiele Osvaldt Rosales
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Camila Fernandes Souza
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Lucas Wenceslau Moreno
- Department of Structural Biology, Molecular and Genetics, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Gustavo Ferreira Alves
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - José Carlos Rebuglio Vellosa
- Department of Clinical and Toxicological Analysis, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Daniel Fernandes
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Jose Rosa Gomes
- Department of Structural Biology, Molecular and Genetics, Post Graduation Program in Biomedical Science, Universidade Estadual de Ponta Grossa, Avenida Carlos Cavalcanti, 4748, Ponta Grossa, PR, 84030-900, Brazil.
| |
Collapse
|
12
|
Liu S, Tan M, Cai J, Li C, Yang M, Sun X, He B. Ribosome-targeting antibiotic control NLRP3-mediated inflammation by inhibiting mitochondrial DNA synthesis. Free Radic Biol Med 2024; 210:75-84. [PMID: 37992790 DOI: 10.1016/j.freeradbiomed.2023.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/08/2023] [Accepted: 11/16/2023] [Indexed: 11/24/2023]
Abstract
While antibiotics are designed to target bacteria specifically, most are known to affect host cell physiology. Certain classes of antibiotics have been reported to have immunosuppressive effects, but the underlying mechanisms remain elusive. Here, we show that doxycycline, a ribosomal-targeting antibiotic, effectively inhibited both mitochondrial translation and nucleotide-binding domain and leucine-rich repeat-containing protein 3 (NLRP3) inflammasome-mediated caspase-1 activation and interleukin-1β (IL-1β) production in bone-marrow-derived macrophages (BMDMs). In addition, knockdown of mitochondrial methionyl-tRNA formyltransferase (Mtfmt), which is rate limiting for mitochondrial translation, also resulted in the inhibition of NLRP3 inflammasome-mediated caspase-1 activation and IL-1β secretion. Furthermore, both doxycycline treatment and Mtfmt knockdown blocked the synthesis of mitochondrial DNA (mtDNA) and the generation of oxidized mtDNA (Ox-mtDNA), which serves as a ligand for NLRP3 inflammasome activation. In addition, in vivo results indicated that doxycycline mitigated NLRP3 inflammasome-dependent inflammation, including lipopolysaccharide-induced systemic inflammation and endometritis. Taken together, the results unveil the antibiotics targeting the mitoribosome have the ability to mitigate NLRP3 inflammasome activation by inhibiting mitochondrial translation and mtDNA synthesis thus opening up new possibilities for the treatment of NLRP3-related diseases.
Collapse
Affiliation(s)
- Suyuan Liu
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Meiling Tan
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Jiangxue Cai
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Chenxuan Li
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Miaoxin Yang
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Xiaoxiao Sun
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Bin He
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, 210095, PR China.
| |
Collapse
|
13
|
Acharya Y, Taneja KK, Haldar J. Dual functional therapeutics: mitigating bacterial infection and associated inflammation. RSC Med Chem 2023; 14:1410-1428. [PMID: 37593575 PMCID: PMC10429821 DOI: 10.1039/d3md00166k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 05/21/2023] [Indexed: 08/19/2023] Open
Abstract
The emergence of antimicrobial resistance, coupled with the occurrence of persistent systemic infections, has already complicated clinical therapy efforts. Moreover, infections are also accompanied by strong inflammatory responses, generated by the host's innate and adaptive immune systems. The closely intertwined relationship between bacterial infection and inflammation has multiple implications on the ability of antibacterial therapeutics to tackle infection and inflammation. Particularly, uncontrolled inflammatory responses to infection can lead to sepsis, a life-threatening physiological condition. In this review, we discuss dual-functional antibacterial therapeutics that have potential to be developed for treating inflammation associated with bacterial infections. Immense research is underway that aims to develop new therapeutic agents that, when administered, regulate the excess inflammatory response, i.e. they have immunomodulatory properties along with the desired antibacterial activity. The classes of antibiotics that have immunomodulatory function in addition to antibacterial activity have been reviewed. Host defense peptides and their synthetic mimics are amongst the most sought-after solutions to develop such dual-functional therapeutics. This review also highlights the important classes of peptidomimetics that exhibit both antibacterial and immunomodulatory properties.
Collapse
Affiliation(s)
- Yash Acharya
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
| | - Kashish Kumar Taneja
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
| |
Collapse
|
14
|
Tauber SC, Nau R. Treatment of septic encephalopathy and encephalitis - a critical appraisal. Expert Rev Neurother 2023; 23:1069-1080. [PMID: 38019041 DOI: 10.1080/14737175.2023.2288652] [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: 09/19/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023]
Abstract
INTRODUCTION The central nervous system is frequently involved during severe sepsis. Patients either develop septic encephalopathy characterized by delirium and coma or focal neurological signs as a consequence of septic-embolic or septic-metastatic encephalitis. AREAS COVERED In this review, a summary of currently available literature on established and some promising experimental treatment options for septic encephalopathy and encephalitis is provided, with a focus on the clinical utility of published studies. EXPERT OPINION Treatment relies on proper identification of the causative pathogen and rapidly initiated adequate empirical or (after identification of the pathogen) tailored antibiotic therapy, fluid and electrolyte management. In the presence of brain abscess(es) or mycotic aneurysm(s), surgery or interventional neuroradiology must be considered. Pharmacological approaches to prevent delirium of different etiology include the use of dexmedetomidine and (with limitations) of melatonin and its derivatives. In the absence of a specific pharmacological treatment, non-pharmacological bundles of interventions (e.g. promotion of sleep, cognitive stimulation, early mobilization and adequate therapy of pain) are of proven efficacy to prevent delirium of different etiology including sepsis. Experimental promising therapies include the use of non-bacteriolytic antibiotics and the reduction of the toxic effects of microglial activation.
Collapse
Affiliation(s)
- Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Aachen, Germany
| | - Roland Nau
- Department of Neuropathology, University Medicine Göttingen, Georg-August-University Göttingen, Göttingen, Germany
- Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| |
Collapse
|
15
|
Pan Y, Chen YX, Zhang J, Lin ML, Liu GM, Xu XL, Fan XQ, Zhong Y, Li Q, Ai SM, Xu W, Tan J, Zhou HF, Xu DD, Zhang HY, Xu B, Wang S, Ma JJ, Zhang S, Gan LY, Cui JT, Li L, Xie YY, Guo X, Pan-Doh N, Zhu ZT, Lu Y, Shi YX, Xia YW, Li ZY, Liang D. Doxycycline vs Placebo at 12 Weeks in Patients With Mild Thyroid-Associated Ophthalmopathy: A Randomized Clinical Trial. JAMA Ophthalmol 2022; 140:1076-1083. [PMID: 36173609 PMCID: PMC9523551 DOI: 10.1001/jamaophthalmol.2022.3779] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/03/2022] [Indexed: 12/15/2022]
Abstract
Importance Mild thyroid-associated ophthalmopathy (TAO) negatively impacts quality of life, yet no clinical guidelines for its treatment are available. Existing evidence supports the use of doxycycline in treating mild TAO. Objective To evaluate the short-term (12 weeks) efficacy of doxycycline in treating mild TAO. Design, Setting, and Participants In this placebo-controlled multicenter randomized double-masked trial, 148 patients were assessed for eligibility. After exclusions (patients who were pregnant or lactating, had an allergy to tetracyclines, or had uncontrolled systematic diseases), 100 patients with mild TAO (orbital soft tissue affected mildly) at 5 centers in China were enrolled from July 2013 to December 2019 and monitored for 12 weeks. Interventions Participants were randomly assigned 1:1 to receive doxycycline (50 mg) or placebo once daily for 12 weeks. Main Outcomes and Measures The primary outcome was the rate of improvement at 12 weeks compared with baseline assessed by a composite indicator of eyelid aperture (reduction ≥2 mm), proptosis (reduction ≥2 mm), ocular motility (increase ≥8°), and Graves ophthalmopathy-specific quality-of-life (GO-QOL) scale score (increase ≥6 points). Adverse events were recorded. Results A total of 50 participants were assigned to doxycycline and 50 to placebo. The mean (SD) age was 36.7 (9.1) years; 75 participants (75.0%) were female and 100 (100.0%) were Asian. Medication compliance was checked during participant interviews and by counting excess tablets. At week 12, the improvement rate was 38.0% (19 of 50) in the doxycycline group and 16.0% (8 of 50) in the placebo group (difference, 22.0%; 95% CI, 5.0-39.0; P = .01) in the intention-to-treat population. The per-protocol sensitivity analysis showed similar results (39.6% [19 of 48] vs 16.0% [8 of 50]; difference, 23.6%; 95% CI, 6.4-40.8; P = .009). No adverse events other than 1 case of mild gastric acid regurgitation was recorded in either group. Conclusions and Relevance The results of this study indicate that oral doxycycline, 50 mg daily, resulted in greater improvement of TAO-related symptoms at 12 weeks compared with placebo in patients with mild TAO. These findings support the consideration of doxycycline for mild TAO but should be tempered by recognizing the relatively short follow-up and the size of the cohort. Trial Registration ClinicalTrials.gov Identifier: NCT02203682.
Collapse
Affiliation(s)
- Yuan Pan
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Yu-Xi Chen
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Jian Zhang
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Miao-Li Lin
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
- C-MER Dennis Lam Eye Hospital, Shenzhen, China
| | - Guang-Ming Liu
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
- Department of Ophthalmology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xue-Liang Xu
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Department of Ophthalmology, Xiangya Changde Hospital, Changde, China
- Hunan Key Laboratory of Ophthalmology, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xian-Qun Fan
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Zhong
- Department of Ophthalmology, Peking Union Medical College Hospital, Beijing, China
| | - Qing Li
- Department of Ophthalmology, Fujian Provincial Hospital, Fuzhou, China
| | - Si-Ming Ai
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Wen Xu
- Department of Endocrinology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jia Tan
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hui-Fang Zhou
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong-Dong Xu
- Department of Ophthalmology, Peking Union Medical College Hospital, Beijing, China
| | - Hui-Ying Zhang
- Department of Ophthalmology, Fujian Provincial Hospital, Fuzhou, China
| | - Bei Xu
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Sha Wang
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jun-Jie Ma
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Ophthalmology, Huzhou Central Hospital, Huzhou, China
| | - Shuo Zhang
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin-Yang Gan
- Department of Ophthalmology, Peking Union Medical College Hospital, Beijing, China
| | - Jian-Tao Cui
- Department of Ophthalmology, Peking Union Medical College Hospital, Beijing, China
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Li Li
- Department of Ophthalmology, Fujian Provincial Hospital, Fuzhou, China
| | - Yan-Yan Xie
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Xinxing Guo
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Dana Center of Preventive Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nathan Pan-Doh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zhuo-Ting Zhu
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Yao Lu
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Yu-Xun Shi
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Yi-Wen Xia
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Zuo-Yi Li
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Dan Liang
- Department of Ocular Immunology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| |
Collapse
|
16
|
Chan D, Ooi E, Khalid O. A systematic review and meta-analysis of the role of doxycycline in chronic rhinosinusitis. J Laryngol Otol 2022; 137:474-483. [PMID: 35919933 DOI: 10.1017/s0022215122001803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The objective of this systematic review and meta-analysis was to evaluate the role of doxycycline in the management of chronic rhinosinusitis. METHOD This was a systematic review using Ovid Medline, Cinahl, Scopus and Cochrane and was limited to meta-analyses, systematic reviews and randomised, clinical trials. A combination of the following search terms was used: 'sinusitis', 'nasal polyps', 'doxycycline' and 'tetracycline'. Raw means and standard deviations were extracted from the included studies. The meta-analysis was performed using mean differences of pre- versus post-doxycycline treatment. RESULTS A total of 279 studies were screened, of which 5 studies met the criteria (all randomised, controlled trials published between 2010 and 2021). The interventions, endpoints and measured outcomes varied across all studies. Meta-analysis performed on pre- versus post-doxycycline treatment for Sino-Nasal Outcome Test-22, nasal polyp scores and symptom scores did not yield statistically significant results. CONCLUSION This review identified a small number of high-quality studies on the use of doxycycline in chronic rhinosinusitis. There does not seem to be convincing evidence for the routine use of doxycycline in patients with chronic rhinosinusitis. Further research may try to identify certain phenotypes of chronic rhinosinusitis that may better respond to doxycycline.
Collapse
Affiliation(s)
- D Chan
- Department of Otorhinolaryngology, Flinders Medical Centre, Adelaide, Australia
| | - E Ooi
- Department of Otorhinolaryngology, Flinders Medical Centre, Adelaide, Australia
| | - O Khalid
- Department of Otorhinolaryngology, Flinders Medical Centre, Adelaide, Australia
| |
Collapse
|
17
|
Preethy S, Raghavan K, Dedeepiya VD, Surya Prakash V, Ikewaki N, Ikeue Y, Nagataki M, Iwasaki M, Senthilkumar R, Abraham SJK. Beneficial Immune Regulation by Biological Response Modifier Glucans in COVID-19 and Their Envisaged Potentials in the Management of Sepsis. Front Immunol 2022; 13:870632. [PMID: 35833122 PMCID: PMC9272021 DOI: 10.3389/fimmu.2022.870632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/23/2022] [Indexed: 12/24/2022] Open
Abstract
Sepsis is a life-threatening condition caused by an abnormal immune response induced by infection with no approved or specific therapeutic options. We present our perspectives for the therapeutic management of sepsis through a four-way approach: (1) infection control through immune enhancement; (2) immune suppression during the initial hyper-inflammatory phase; (3) balanced immune-modulation to counter the later immune-paralysis phase; and (4) advantageous effects on metabolic and coagulation parameters throughout. COVID-19 is a virus-triggered, accelerated sepsis-like reaction that is associated with the rapid progress of an inflammatory cascade involving a cytokine storm and multiorgan failure. Here, we discuss the potential of the biological response modifiers, β-glucans (BRMGs), in the management of sepsis based on their beneficial effects on inflammatory-immune events in COVID-19 clinical studies. In COVID-19 patients, apart from metabolic regulation, BRMGs, derived from a black yeast, Aureobasidium pullulans strain AFO-202, have been reported to stimulate immune responses. BRMGs, produced by another strain (N-163) of A. pullulans, have been implicated in the beneficial regulation of inflammatory markers and immunity, namely IL-6, C-reactive protein (CRP), D-Dimer, ferritin, neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-C-reactive protein ratio (LCR), leucocyte-to-C-reactive protein ratio (LeCR), and leukocyte-to-IL-6 ratio (LeIR). Agents such as these β-glucans, which are safe as they have been widely consumed by humans for decades, have potential as adjuncts for the prevention and management of sepsis as they exert their beneficial effects across the spectrum of processes and factors involved in sepsis pathology, including, but not limited to, metabolism, infection, inflammation, immune modulation, immune enhancement, and gut microbiota.
Collapse
Affiliation(s)
- Senthilkumar Preethy
- Fujio-Eiji Academic Terrain (FEAT), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India
| | - Kadalraja Raghavan
- Department of Paediatric Neurology, Sarvee Integra Private Limited, Chennai, India
- Department of Paediatric Neurology, Jesuit Antonyraj memorial Inter-disciplinary Centre for Advanced Recovery and Education (JAICARE), Madurai, India
| | | | | | - Nobunao Ikewaki
- Department of Medical Life Science, Kyushu University of Health and Welfare, Nobeoka, Japan
- Institute of Immunology, Junsei Educational Institute, Nobeoka, Japan
| | | | | | - Masaru Iwasaki
- Centre for Advancing Clinical Research (CACR), University of Yamanashi - School of Medicine, Chuo, Japan
| | - Rajappa Senthilkumar
- Fujio-Eiji Academic Terrain (FEAT), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India
| | - Samuel J. K. Abraham
- Mary-Yoshio Translational Hexagon (MYTH), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India
- Centre for Advancing Clinical Research (CACR), University of Yamanashi - School of Medicine, Chuo, Japan
- Antony-Xavier Interdisciplinary Scholastics (AXIS), GN Corporation Ltd., Kofu, Japan
| |
Collapse
|
18
|
Sauer A, Putensen C, Bode C. Immunomodulation by Tetracyclines in the Critically Ill: An Emerging Treatment Option? Crit Care 2022; 26:74. [PMID: 35337355 PMCID: PMC8951664 DOI: 10.1186/s13054-022-03909-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
This article is one of ten reviews selected from the Annual Update in Intensive Care and Emergency Medicine 2022. Other selected articles can be found online at https://www.biomedcentral.com/collections/annualupdate2022 . Further information about the Annual Update in Intensive Care and Emergency Medicine is available from https://link.springer.com/bookseries/8901 .
Collapse
Affiliation(s)
- Andrea Sauer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Putensen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
19
|
Combined Therapy with Ivermectin and Doxycycline can effectively alleviate the Cytokine Storm of COVID-19 Infection amid Vaccination Drive: A Narrative Review. J Infect Public Health 2022; 15:566-572. [PMID: 35462191 PMCID: PMC8964533 DOI: 10.1016/j.jiph.2022.03.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/22/2022] Open
Abstract
An unprecedented global health crisis has developed due to the emergence of the mysterious coronavirus-2 of the severe acute respiratory syndrome, which has resulted in millions of deaths around the globe, as no therapy could control the ‘cytokine storm’. Consequently, many vaccines have been developed and several others are being developed for this infection. Although most of the approved vaccines have been highly effective, many developing, and economically poor countries are still deprived of vaccination against SARS-CoV-2 due to the unequal distribution of vaccines worldwide. Furthermore, the uncertainty about the effectiveness of the available vaccines against the emerging mutants and variants also remains a matter of concern. Due to the multistep pathogenesis and unique features, combination therapy using safe immunomodulatory and antiviral drugs should be considered as the most effective and acceptable therapeutic regimen for this infection. Based on a thorough assessment of the literature, it was determined that it would be interesting to study the therapeutic potential of ivermectin and doxycycline, given their roles in several biological pathways involved in SARS CoV-2 pathogenesis. Following that, a comprehensive literature search was undertaken using Scopus, Web of Science, and Pubmed, depending on the inclusion and exclusion criteria. The present study provides a mechanism and comprehensive report, highlighting the role of combined therapy with ivermectin and doxycycline in alleviating the ‘cytokine storm’ of COVID-19 infection.
Collapse
|
20
|
Effect of Doxycycline in Decreasing the Severity of Clostridioides difficile Infection in Mice. Antibiotics (Basel) 2022; 11:antibiotics11010116. [PMID: 35052993 PMCID: PMC8772929 DOI: 10.3390/antibiotics11010116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 11/28/2022] Open
Abstract
Background: Doxycycline possesses antibacterial activity against Clostridioides difficile and anti-inflammatory effects. Materials and Methods: The influence of doxycycline on the development of CDI was studied in an established animal model of CDI using C57BL/6 mice. Results: Mice intraperitoneally administered doxycycline had higher cecum weight (1.3 ± 0.1 vs. 0.5 ± 0.1 g; p < 0.001) and less body weight reduction (0.7 ± 0.5 g vs. −17.4 ± 0.2 g; p < 0.001) than untreated mice infected with C. difficile. Oral doxycycline, metronidazole, or vancomycin therapy resulted in less body weight reduction in mice with CDI than in untreated mice (1.1 ± 0.1 g, 1.3 ± 0.2 g, 1.2 ± 0.1 g, vs. 2.9 ± 0.3 g; p < 0.001). Doxycycline therapy led to lower expression levels of inflammatory cytokines, such as macrophage inflammatory protein-2 (0.4 ± 0.1 vs. 2.9 ± 1.3, p = 0.02), and higher levels of zonula occludens-1 (1.2 ± 0.1 vs. 0.8 ± 0.1, p = 0.02) in colonic tissues than in untreated mice. Conclusions: Concurrent intraperitoneal administration of doxycycline and oral C. difficile challenge does not aggravate the disease severity of CDI, and oral doxycycline may be a potential therapeutic option for CDI.
Collapse
|
21
|
Sauer A, Peukert K, Putensen C, Bode C. Antibiotics as immunomodulators: a potential pharmacologic approach for ARDS treatment. Eur Respir Rev 2021; 30:210093. [PMID: 34615700 PMCID: PMC9489085 DOI: 10.1183/16000617.0093-2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/02/2021] [Indexed: 11/05/2022] Open
Abstract
First described in the mid-1960s, acute respiratory distress syndrome (ARDS) is a life-threatening form of respiratory failure with an overall mortality rate of approximately 40%. Despite significant advances in the understanding and treatment of ARDS, no substantive pharmacologic therapy has proven to be beneficial, and current management continues to be primarily supportive. Beyond their antibacterial activity, several antibiotics such as macrolides and tetracyclines exert pleiotropic immunomodulatory effects that might be able to rectify the dysregulated inflammatory response present in patients with ARDS. This review aims to provide an overview of preclinical and clinical studies that describe the immunomodulatory effects of antibiotics in ARDS. Moreover, the underlying mechanisms of their immunomodulatory properties will be discussed. Further studies are necessary to investigate their full therapeutic potential and to identify ARDS phenotypes which are most likely to benefit from their immunomodulatory effects.
Collapse
Affiliation(s)
- Andrea Sauer
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Konrad Peukert
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Putensen
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Bode
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
22
|
Kan X, Chen Y, Huang B, Fu S, Guo W, Ran X, Cao Y, Xu D, Cheng J, Yang Z, Xu Y. Effect of Palrnatine on lipopolysaccharide-induced acute lung injury by inhibiting activation of the Akt/NF -κB pathway. J Zhejiang Univ Sci B 2021; 22:929-940. [PMID: 34783223 DOI: 10.1631/jzus.b2000583] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Inflammation plays an important role in the development of acute lung injury (ALI). Severe pulmonary inflammation can cause acute respiratory distress syndrome (ARDS) or even death. Expression of proinflammatory interleukin-1β (IL-1β) and inducible nitric oxide synthase (iNOS) in the process of pulmonary inflammation will further exacerbate the severity of ALI. The purpose of this study was to explore the effect of Palrnatine (Pa) on lipopolysaccharide (LPS)-induced mouse ALI and its underlying mechanism. Pa, a natural product, has a wide range of pharmacological activities with the potential to protect against lung injury. Western blotting and quantitative real-time polymerase chain reaction (qRT-PCR) assays were performed to detect the expression and translation of inflammatory genes and proteins in vitro and in vivo. Immunoprecipitation was used to detect the degree of P65 translocation into the nucleus. We also used molecular modeling to further clarify the mechanism of action. The results showed that Pa pretreatment could significantly inhibit the expression and secretion of the inflammatory cytokine IL-1β, and significantly reduce the protein level of the proinflammatory protease iNOS, in both in vivo and in vitro models induced by LPS. Further mechanism studies showed that Pa could significantly inhibit the activation of the protein kinase B (Akt)/nuclear factor-κB (NF-κB) signaling pathway in the LPS-induced ALI mode and in LPS-induced RAW264.7 cells. Through molecular dynamics simulation, we observed that Pa was bound to the catalytic pocket of Akt and effectively inhibited the biological activity of Akt. These results indicated that Pa significantly relieves LPS-induced ALI by activating the Akt/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xingchi Kan
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Yingsheng Chen
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Bingxu Huang
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Shoupeng Fu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Wenjin Guo
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Xin Ran
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Yu Cao
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Dianwen Xu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Ji Cheng
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Zhanqing Yang
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Yanling Xu
- Department of Respiratory Medicine, the Second Hospital, Jilin University, Changchun 130012, China.
| |
Collapse
|
23
|
Usmani J, Khan T, Ahmad R, Sharma M. Potential role of herbal medicines as a novel approach in sepsis treatment. Biomed Pharmacother 2021; 144:112337. [PMID: 34688080 DOI: 10.1016/j.biopha.2021.112337] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/09/2021] [Accepted: 10/11/2021] [Indexed: 12/17/2022] Open
Abstract
The growing number of deaths related to sepsis has become a major concern for past few years. Sepsis is a complex pathological reactions that is explained by series of host response to microbial insult. The resulted systemic reactions are manifested by early appearance of proinflammatory cytokines leading to hyperinflammatory phase which is followed by septic shock and death of the patient. The present study has revealed that antibiotics are not self-sufficient to control the complex mechanism of sepsis. Moreover prolonged and unnecessary administration of antibiotics may lead to antibiotic resistance to pathogens. In addition to this, immunosuppressive medications are selective and have targeted approach to certain study population. Drugs from herbal origin have shown to possess a mammoth of immunomodulatory potential by suppressing proinflammatory and anti-inflammatory cytokines exhibiting no or minimal unwanted secondary responses. Concomitantly, herbal plants tend to modulate oxidative stress level and haematological imbalance during inflammatory diseased conditions. Natural compounds have gained much attention for the treatment of several clinical complications. Considering the promising responses of medicinal plants with less/no side effects and easy procurement, comprehensive research on herbal plants to treat sepsis should be contemplated.
Collapse
Affiliation(s)
- Juveria Usmani
- Department of Pharmacology, School of Pharmaceutical Sciences & Research, Jamia Hamdard, New Delhi, India
| | - Tahira Khan
- Department of Pharmacology, School of Pharmaceutical Sciences & Research, Jamia Hamdard, New Delhi, India
| | - Razi Ahmad
- Department of Pharmacology, Hamdard Institute of Medical Sciences & Research, Jamia Hamdard, New Delhi 110019, India.
| | - Manju Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences & Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
24
|
Wu M, Lai T, Jing D, Yang S, Wu Y, Li Z, Wu Y, Zhao Y, Zhou L, Chen H, Shen J, Li W, Ying S, Chen Z, Wu X, Shen H. Epithelium-derived IL17A Promotes Cigarette Smoke-induced Inflammation and Mucus Hyperproduction. Am J Respir Cell Mol Biol 2021; 65:581-592. [PMID: 34186014 DOI: 10.1165/rcmb.2020-0424oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Airway epithelium is a central modulator of innate and adaptive immunity in the lung. Interleukin (IL)17A expression was found to be increased in airway epithelium; however, the role of epithelial-derived IL17A in chronic obstructive pulmonary disease (COPD) remains unclear. In this study, we aim to determine whether epithelial-derived IL17A regulates inflammation and mucus hyperproduction in COPD using a cultured human bronchial epithelial (HBE) cell line in vitro and airway epithelium IL17A-specific knockout mouse in vivo. Increased IL17A expression was observed in mouse airway epithelium upon cigarette smoke (CS) exposure or in a COPD mouse model that was induced by CS and elastin. CS extract (CSE) also triggered IL17A expression in HBE cells. Blocking IL17A or IL17RA effectively attenuated CSE-induced MUC5AC and the inflammatory cytokines IL6, tumor necrosis factor (TNF)-α, and IL1β in HBE cells, suggesting that IL17A mediates CSE-induced inflammation and mucin production in an autocrine manner. CSE activated p-JUN and p-JNK, which were also reduced by IL17RA-siRNA, and JUN-siRNA attenuated CSE-induced IL6 and MUC5AC. In vivo, selective knockout of IL17A in airway epithelium markedly reduced the neutrophilic infiltration in Bronchoalveolar Lavage Fluid (BALF), peribronchial inflammation, pro-inflammatory mediators (CXCL1 and CXCL2), and mucus production in a COPD mouse model. We showed a novel function of airway epithelium-derived IL17A, which can act locally in an autocrine manner to amplify inflammation and increase mucus production in COPD pathogenesis.
Collapse
Affiliation(s)
- Mindan Wu
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Tianwen Lai
- Zhejiang University School of Medicine, 26441, Hangzhou, China
| | - Du Jing
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Shiyi Yang
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Yanping Wu
- Zhejiang University School of Medicine, 26441, Respiratory and Critical Care Medicine, Hangzhou, China
| | - Zhouyang Li
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Yinfang Wu
- Zhejiang University School of Medicine, 26441, Respiratory and Critical Care Medicine, Hangzhou, China
| | - Yun Zhao
- Zhejiang University School of Medicine, 26441, Respiratory and Critical Care Medicine, Hangzhou, China
| | - Lingren Zhou
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Haipin Chen
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Jiaxin Shen
- Zhejiang University School of Medicine, 26441, Respiratory and Critical Care Medicine, Hangzhou, China
| | - Wen Li
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Respiratory and Critical Care Midicine, Hangzhou, China
| | - Songmin Ying
- Zhejiang University School of Medicine, 26441, Respiratory and Critical Care Medicine, Hangzhou, China
| | - Zhihua Chen
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Respiratory and Critical Care Midicine, Hangzhou, China
| | - Xiaohong Wu
- Zhejiang University School of Medicine Sir Run Run Shaw Hospital, 56660, Hangzhou, China
| | - Huahao Shen
- Zhejiang University School of Medicine, 26441, Respiratory Medicine, Hangzhou, China;
| |
Collapse
|
25
|
Al-kuraishy HM, Al-Gareeb AI, Alqarni M, Cruz-Martins N, El-Saber Batiha G. Pleiotropic Effects of Tetracyclines in the Management of COVID-19: Emerging Perspectives. Front Pharmacol 2021; 12:642822. [PMID: 33967777 PMCID: PMC8103613 DOI: 10.3389/fphar.2021.642822] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/18/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a global infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Approximately 15% of severe cases require an intensive care unit (ICU) admission and mechanical ventilation due to development of acute respiratory distress syndrome (ARDS). Tetracyclines (TCs) are a group of bacteriostatic antibiotics, like tetracycline, minocycline, and doxycycline, effective against aerobic and anaerobic bacteria as well as Gram-positive and Gram-negative bacteria. Based on available evidences, TCs may be effective against coronaviruses and thus useful to treat COVID-19. Thus, this review aims to provide a brief overview on the uses of TCs for COVID-19 management. SARS-CoV-2 and other coronaviruses depend mainly on the matrix metalloproteinases (MMPs) for their proliferation, cell adhesion, and infiltration. The anti-inflammatory mechanisms of TCs are linked to different pathways. Briefly, TCs inhibit mitochondrial cytochrome c and caspase pathway with improvement of lymphopenia in early COVID-19. Specifically, minocycline is effective in reducing COVID-19-related complications, through attenuation of cytokine storm as apparent by reduction of interleukin (IL)-6, IL-1, and tumor necrosis factor (TNF)-α. Different clinical trials recommend the replacement of azithromycin by minocycline in the management of COVID-19 patients at high risk due to two main reasons: 1) minocycline does not prolong the QT interval and even inhibits ischemia-induced arrhythmia; 2) minocycline displays synergistic effect with chloroquine against SARS-CoV-2. Taken together, the data presented here show that TCs, mainly doxycycline or minocycline, may be potential partners in COVID-19 management, derived pneumonia, and related complications, such as acute lung injury (ALI) and ARDS.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, BaghdadIraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, BaghdadIraq
| | - Mohammed Alqarni
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Porto, Portugal
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
26
|
Park CS, Kim SH, Lee CK. Immunotherapy of Autoimmune Diseases with Nonantibiotic Properties of Tetracyclines. Immune Netw 2020; 20:e47. [PMID: 33425432 PMCID: PMC7779869 DOI: 10.4110/in.2020.20.e47] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/08/2020] [Indexed: 01/12/2023] Open
Abstract
Tetracyclines, which have long been used as broad-spectrum antibiotics, also exhibit a variety of nonantibiotic activities including anti-inflammatory and immunomodulatory properties. Tetracyclines bind to the 30S ribosome of the bacteria and inhibit protein synthesis. Unlike antimicrobial activity, the primary molecular target for the nonantibiotic activity of tetracycline remains to be clarified. Nonetheless, the therapeutic efficacies of tetracyclines, particularly minocycline and doxycycline, have been demonstrated in various animal models of autoimmune disorders, such as multiple sclerosis, rheumatoid arthritis, and asthma. In this study, we summarized the anti-inflammatory and immunomodulatory activities of tetracyclines, focusing on the mechanisms underlying these activities. In addition, we highlighted the on-going or completed clinical trials with reported outcomes.
Collapse
Affiliation(s)
- Chan-Su Park
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sang-Hyun Kim
- Department of Pharmaceutics, College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Chong-Kil Lee
- Department of Pharmaceutics, College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
27
|
Gendrot M, Andreani J, Jardot P, Hutter S, Delandre O, Boxberger M, Mosnier J, Le Bideau M, Duflot I, Fonta I, Rolland C, Bogreau H, La Scola B, Pradines B. In Vitro Antiviral Activity of Doxycycline against SARS-CoV-2. Molecules 2020; 25:E5064. [PMID: 33142770 PMCID: PMC7663271 DOI: 10.3390/molecules25215064] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/22/2020] [Accepted: 10/30/2020] [Indexed: 01/18/2023] Open
Abstract
In December 2019, a new severe acute respiratory syndrome coronavirus (SARS-CoV-2), causing coronavirus disease 2019 (COVID-19), emerged in Wuhan, China. Despite containment measures, SARS-CoV-2 spread in Asia, Southern Europe, then in America and currently in Africa. Identifying effective antiviral drugs is urgently needed. An efficient approach to drug discovery is to evaluate whether existing approved drugs can be efficient against SARS-CoV-2. Doxycycline, which is a second-generation tetracycline with broad-spectrum antimicrobial, antimalarial and anti-inflammatory activities, showed in vitro activity on Vero E6 cells infected with a clinically isolated SARS-CoV-2 strain (IHUMI-3) with median effective concentration (EC50) of 4.5 ± 2.9 µM, compatible with oral uptake and intravenous administrations. Doxycycline interacted both on SARS-CoV-2 entry and in replication after virus entry. Besides its in vitro antiviral activity against SARS-CoV-2, doxycycline has anti-inflammatory effects by decreasing the expression of various pro-inflammatory cytokines and could prevent co-infections and superinfections due to broad-spectrum antimicrobial activity. Therefore, doxycycline could be a potential partner of COVID-19 therapies. However, these results must be taken with caution regarding the potential use in SARS-CoV-2-infected patients: it is difficult to translate in vitro study results to actual clinical treatment in patients. In vivo evaluation in animal experimental models is required to confirm the antiviral effects of doxycycline on SARS-CoV-2 and more trials of high-risk patients with moderate to severe COVID-19 infections must be initiated.
Collapse
Affiliation(s)
- Mathieu Gendrot
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 13005 Marseille, France; (M.G.); (O.D.); (J.M.); (I.F.); (H.B.)
- Aix Marseille Univ, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France;
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
| | - Julien Andreani
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
- Aix Marseille Univ, IRD, AP-HM, MEPHI, 13005 Marseille, France
| | - Priscilla Jardot
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
- Aix Marseille Univ, IRD, AP-HM, MEPHI, 13005 Marseille, France
| | - Sébastien Hutter
- Aix Marseille Univ, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France;
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
| | - Océane Delandre
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 13005 Marseille, France; (M.G.); (O.D.); (J.M.); (I.F.); (H.B.)
- Aix Marseille Univ, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France;
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
| | - Manon Boxberger
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
- Aix Marseille Univ, IRD, AP-HM, MEPHI, 13005 Marseille, France
| | - Joel Mosnier
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 13005 Marseille, France; (M.G.); (O.D.); (J.M.); (I.F.); (H.B.)
- Aix Marseille Univ, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France;
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
- Centre National de Référence du Paludisme, 13005 Marseille, France
| | - Marion Le Bideau
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
- Aix Marseille Univ, IRD, AP-HM, MEPHI, 13005 Marseille, France
| | - Isabelle Duflot
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
- Aix Marseille Univ, IRD, AP-HM, MEPHI, 13005 Marseille, France
| | - Isabelle Fonta
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 13005 Marseille, France; (M.G.); (O.D.); (J.M.); (I.F.); (H.B.)
- Aix Marseille Univ, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France;
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
- Centre National de Référence du Paludisme, 13005 Marseille, France
| | - Clara Rolland
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
- Aix Marseille Univ, IRD, AP-HM, MEPHI, 13005 Marseille, France
| | - Hervé Bogreau
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 13005 Marseille, France; (M.G.); (O.D.); (J.M.); (I.F.); (H.B.)
- Aix Marseille Univ, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France;
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
- Centre National de Référence du Paludisme, 13005 Marseille, France
| | - Bernard La Scola
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
- Aix Marseille Univ, IRD, AP-HM, MEPHI, 13005 Marseille, France
| | - Bruno Pradines
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 13005 Marseille, France; (M.G.); (O.D.); (J.M.); (I.F.); (H.B.)
- Aix Marseille Univ, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France;
- IHU Méditerranée Infection, 13005 Marseille, France; (J.A.); (P.J.); (M.B.); (M.L.B.); (I.D.); (C.R.)
- Centre National de Référence du Paludisme, 13005 Marseille, France
| |
Collapse
|