1
|
Singh S, Gumbo T, Wang JY, Boorgula GD, Burke A, Huang HL, McShane PJ, Amaro-Galvez R, Gross JE, Aryal S, Heysell SK, Srivastava S. Imipenem pharmacokinetics/pharmacodynamics in preclinical hollow fiber model, dose-finding in virtual patients, and clinical evidence of efficacy for Mycobacterium abscessus lung disease. J Infect Dis 2024:jiae601. [PMID: 39688388 DOI: 10.1093/infdis/jiae601] [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: 08/20/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Guideline-based therapy (GBT) for Mycobacterium abscessus (Mab) lung disease achieves sputum culture conversion rates (SCC) of 35%. This poor GBT efficacy is mirrored in the hollow fiber system model of Mab (HFS-Mab). While imipenem is part of GBT, biological effect with or without β-lactamase inhibitors, is unproven. METHODS We performed imipenem/relebactam minimum inhibitory concentration (MIC) in 122 Mab isolates, and an exposure-response study in the HFS-Mab using human intrapulmonary pharmacokinetics. The % time that concentration persisted above MIC (TMIC) mediating maximal effect in the HFS-Mab was used as the exposure target in 10,000 virtual subjects Monte Carlo experiment (MCE)-based dose-finding. For real-world evidence, we performed a patient, intervention (imipenem), comparison (no β-lactam), and outcome (SCC) (PICO) analysis. RESULTS Imipenem killed 1.32 log10 CFU/mL below day 0 in HFS-Mab. Imipenem target exposure was TMIC=47.9±9.77%. 1g infusion, every 6h, achieved the target in >90% of virtual patients in MCEs. The pharmacokinetics/pharmacodynamics MIC breakpoint was 1mg/L. In PICO analyses, median days-to-SCC were 470 in comparators, 311 for imipenem added on failing regimen, and 37 in newly treated (p=0.049). The odds ratio for SCC when imipenem was part of the initial regimen versus comparators was 12.5 (95% confidence interval: 1.47 to 84.55). Patients on imipenem experienced no treatment-limiting adverse event, while 2/7 comparators did (p=0.0457). Middlebrook 7H9 broth MIC distribution, read at 24 hours, correlated better with patient responses than cation-adjusted Mueller Hinton broth. CONCLUSION Imipenem demonstrated biologic effect in the HFS-Mab and in patients. Imipenem/relebactam doses of 1g every 6h are recommended.
Collapse
Affiliation(s)
- Sanjay Singh
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Texas at Tyler, Tyler, Texas, USA
| | - Tawanda Gumbo
- Mathematical Modeling and AI Department, Praedicare Inc., Dallas, Texas, USA
- Hollow Fiber System & Experimental Therapeutics Laboratories, Wet Lab Systems, Praedicare Inc., Dallas, Texas, USA
| | - Jann-Yuan Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Gunavanthi D Boorgula
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Texas at Tyler, Tyler, Texas, USA
| | - Andrew Burke
- The Prince Charles Hospital Rode Rd Chermside, Queensland, Australia
| | - Hung-Ling Huang
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Taiwan
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Taiwan
| | - Pamela J McShane
- Department of Medicine, Section of Pulmonary and Critical Care, School of Medicine, University of Texas at Tyler, Tyler, Texas, USA
| | - Rodolfo Amaro-Galvez
- Department of Medicine, Section of Pulmonary and Critical Care, School of Medicine, University of Texas at Tyler, Tyler, Texas, USA
| | | | - Santosh Aryal
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, Texas, USA
| | - Scott K Heysell
- Division of Infectious Diseases and International Health, University of Virginia, USA
| | - Shashikant Srivastava
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Texas at Tyler, Tyler, Texas, USA
- Department of Cellular and Molecular Biology, University of Texas Health Science Centre at Tyler, Tyler, Texas, USA
| |
Collapse
|
2
|
Su J, Lin C, Lin X, Hu S, Deng X, Xie L, Ye H, Zhou F, Wu S. Combining ulinastatin with TIENAM improves the outcome of sepsis induced by cecal ligation and puncture in mice by reducing inflammation and regulating immune responses. Int Immunopharmacol 2024; 141:112927. [PMID: 39163689 DOI: 10.1016/j.intimp.2024.112927] [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: 06/19/2024] [Revised: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Despite the high mortality associated with sepsis, effective and targeted treatments remain scarce. The use of conventional antibiotics such as TIENAM (imipenem and cilastatin sodium for injection, TIE) is challenging because of the increasing bacterial resistance, which diminishes their efficacy and leads to adverse effects. Our previous studies demonstrated that ulinastatin (UTI) exerts a therapeutic impact on sepsis by reducing systemic inflammation and modulating immune responses. In this study, we examined the possibility of administering UTI and TIE after inducing sepsis in a mouse model using cecal ligation and puncture (CLP). We assessed the rates of survival, levels of inflammatory cytokines, the extent of tissue damage, populations of immune cells, microbiota in ascites, and important signaling pathways. The combination of UTI and TIE significantly improved survival rates and reduced inflammation and bacterial load in septic mice, indicating potent antimicrobial properties. Notably, the survival rates of UTI+TIE-treated mice increased from 10 % to 75 % within 168 h compared to those of mice that were subjected to CLP. The dual treatment successfully regulated the levels of inflammatory indicators (interleukin [IL]-6, IL-1β, and tumor necrosis factor [TNF]-α) and immune cell numbers by reducing B cells, natural killer cells, and TNFR2+ Treg cells and increasing CD8+ T cells. Additionally, the combination of UTI and TIE alleviated tissue damage, reduced bacterial load in the peritoneal cavity, and suppressed the NF-κB signaling pathway. Our findings indicate that UTI and TIE combination therapy can significantly enhance sepsis outcomes by reducing inflammation and boosting the immune system. The results offer a promising therapeutic approach for future sepsis treatment.
Collapse
Affiliation(s)
- Jingqian Su
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China.
| | - Congfan Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Xinrui Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Shan Hu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Xiaohui Deng
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Lian Xie
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Hui Ye
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Fen Zhou
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Shun Wu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| |
Collapse
|
3
|
Turkeltaub L, Kashat L, Assous MV, Adler K, Bar-Meir M. Estimating bacterial load in S. aureus and E. coli bacteremia using bacterial growth graph from the continuous monitoring blood culture system. Eur J Clin Microbiol Infect Dis 2024; 43:1931-1938. [PMID: 39073670 PMCID: PMC11405537 DOI: 10.1007/s10096-024-04893-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: 04/22/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND We examined whether the time to positivity (TTP) and growth and detection plot graph (GDPG) created by the automated blood culture system can be used to determine the bacterial load in bacteremic patients and its potential association correlation with disease severity. METHODS Known bacterial inocula were injected into the blood culture bottles. The GDPGs for the specific inocula were downloaded and plotted. A cohort of 30 consecutive clinical cultures positive for S. aureus and E. coli was identified. Bacterial load was determined by comparing the GDPG with the "standard" curves. Variables associated with disease severity were compared across 3 bacterial load categories (< 100, 100-1000, > 1000 CFU/mL). RESULTS S. aureus growth was sensitive to the blood volume obtained whereas E. coli growth was less so. A 12-hour delay in sample transfer to the microbiology laboratory resulted in a decrease in TTP by 2-3 h. Mean TTP was 15 and 10 h for S. aureus and E. coli, respectively, which correlates with > 1000 CFU/mL and 500-1000 CFU/ml. For S. aureus, patients with a bacterial load > 100 CFU/mL had a higher mortality rate, (OR for death = 9.7, 95% CI 1.6-59, p = 0.01). Bacterial load > 1000 CFU/mL had an odds ratio of 6.4 (95% CI1.2-35, p = 0.03) to predict an endovascular source. For E. coli bacteremia, we did not find any correlations with disease severity. CONCLUSION GDPG retrieved from the automated blood culture system can be used to estimate bacterial load. S.aureus bacterial load, but not E.coli, was associated with clinical outcome.
Collapse
Affiliation(s)
- Leehe Turkeltaub
- Pediatric Department, Shaare-Zedek Medical Center, Jerusalem, Israel
| | - Livnat Kashat
- The Microbiology Laboratory, Shaare-Zedek Medical Center, Jerusalem, Israel
| | - Marc V Assous
- The Microbiology Laboratory, Shaare-Zedek Medical Center, Jerusalem, Israel.
- The Faculty of Medicine, The Hebrew University, Jerusalem, Israel.
| | - Karen Adler
- The Microbiology Laboratory, Shaare-Zedek Medical Center, Jerusalem, Israel
| | - Maskit Bar-Meir
- Pediatric Infectious Diseases, Shaare-Zedek Medical Center, Jerusalem, Israel.
- The Faculty of Medicine, The Hebrew University, Jerusalem, Israel.
| |
Collapse
|
4
|
Zhang Y, Wu D, Sun Q, Luo Z, Zhang Y, Wang B, Chen W. Atorvastatin combined with imipenem alleviates lung injury in sepsis by inhibiting neutrophil extracellular trap formation via the ERK/NOX2 signaling pathway. Free Radic Biol Med 2024; 220:179-191. [PMID: 38704053 DOI: 10.1016/j.freeradbiomed.2024.05.006] [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: 02/05/2024] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 05/06/2024]
Abstract
Sepsis is a systemic inflammatory response syndrome caused by the invasion of pathogenic microorganisms. Despite major advances in diagnosis and technology, morbidity and mortality remain high. The level of neutrophil extracellular traps (NETs) is closely associated with the progression and prognosis of sepsis, suggesting the regulation of NET formation as a new strategy in sepsis treatment. Owing to its pleiotropic effects, atorvastatin, a clinical lipid-lowering drug, affects various aspects of sepsis-related inflammation and immune responses. To align closely with clinical practice, we combined it with imipenem for the treatment of sepsis. In this study, we used a cecum ligation and puncture-induced lung injury mouse model and employed techniques including western blot, immunofluorescence, and enzyme-linked immunosorbent assay to measure the levels of NETs and other sepsis-related lung injury indicators. Our findings indicate that atorvastatin effectively inhibited the formation of NETs. When combined with imipenem, it significantly alleviated lung injury, reduced systemic inflammation, and improved the 7-day survival rate of septic mice. Additionally, we explored the inhibitory mechanism of atorvastatin on NET formation in vitro, revealing its potential action through the ERK/NOX2 pathway. Therefore, atorvastatin is a potential immunomodulatory agent that may offer new treatment strategies for patients with sepsis in clinical settings.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, The Key Laboratory of Precision Anesthesia & Perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| | - Di Wu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, The Key Laboratory of Precision Anesthesia & Perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| | - Qishun Sun
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, The Key Laboratory of Precision Anesthesia & Perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| | - Zhen Luo
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, The Key Laboratory of Precision Anesthesia & Perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| | - Yuhao Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, The Key Laboratory of Precision Anesthesia & Perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| | - Bowei Wang
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Wenting Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, The Key Laboratory of Precision Anesthesia & Perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
5
|
Viana JPM, Costa FF, Dias TG, Mendes PM, Copeland GB, Nascimento WS, Mendes SSN, Figueiredo IFS, Fernandes ES, Bocca AL, Maciel MCG. Glucans: A Therapeutic Alternative for Sepsis Treatment. J Immunol Res 2024; 2024:6876247. [PMID: 38939744 PMCID: PMC11208795 DOI: 10.1155/2024/6876247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 06/29/2024] Open
Abstract
Sepsis treatment is a challenging condition due to its complexity, which involves host inflammatory responses to a severe and potentially fatal infection, associated with organ dysfunction. The aim of this study was to analyze the scientific literature on the immunomodulatory effects of glucans in a murine model of systemic infection induced by cecal ligation and puncture. This study comprises an integrative literature review based on systematic steps, with searches carried out in the PubMed, ScienceDirect, Scopus, Web of Science, and Embase databases. In most studies, the main type of glucan investigated was β-glucan, at 50 mg/kg, and a reduction of inflammatory responses was identified, minimizing the occurrence of tissue damage leading to increased animal survival. Based on the data obtained and discussed in this review, glucans represent a promising biotechnological alternative to modulate the immune response and could potentially be used in the clinical management of septic individuals.
Collapse
Affiliation(s)
- Jesse P. M. Viana
- Departamento de Biologia CelularInstituto de Ciências BiológicasPrograma de Pós Graduação em Ciências Biológicas (Biologia Molecular)Laboratório de Imunologia AplicadaUniversidade de Brasília (UnB), Brasília, Brazil
| | - Fernanda F. Costa
- Programa de Pós-graduação em Saúde e TecnologiaUniversidade Federal do Maranhão, São Luís, Brazil
| | - Tatielle G. Dias
- Programa de Pós-graduação em Ciências da SaúdeUniversidade Federal do Maranhão, São Luís, Brazil
| | - Priscila M. Mendes
- Programa de Pós-graduação em Ciências da SaúdeUniversidade Federal do Maranhão, São Luís, Brazil
| | - Gabriel B. Copeland
- Laboratório de Imunologia AplicadaUniversidade de Brasília (UnB), Brasília, Brazil
| | | | - Sofia S. N. Mendes
- Laboratório de Imunologia AplicadaUniversidade de Brasília (UnB), Brasília, Brazil
| | - Isabella F. S. Figueiredo
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente—Faculdades Pequeno PríncipeInstituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | - Elizabeth S. Fernandes
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente—Faculdades Pequeno PríncipeInstituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | - Anamelia L. Bocca
- Departamento de Biologia CelularInstituto de Ciências BiológicasPrograma de Pós Graduação em Ciências Biológicas (Biologia Molecular)Laboratório de Imunologia AplicadaUniversidade de Brasília (UnB), Brasília, Brazil
- Plataforma Bi-Institucional de Pesquisa Translacional—Fiocruz/SP, São Paulo, Brazil
| | - Márcia C. G. Maciel
- Departamento de Biologia CelularInstituto de Ciências BiológicasPrograma de Pós Graduação em Ciências Biológicas (Biologia Molecular)Laboratório de Imunologia AplicadaUniversidade de Brasília (UnB), Brasília, Brazil
- Programa de Pós-graduação em Saúde e TecnologiaUniversidade Federal do Maranhão, São Luís, Brazil
- Programa de Pós-graduação em Ciências da SaúdeUniversidade Federal do Maranhão, São Luís, Brazil
| |
Collapse
|
6
|
Li L, Jiang L, Mao S, Ye J. TLR9 Knockdown Alleviates Sepsis via Disruption of MyD88/NF-κB Pathway Activation. Crit Rev Immunol 2024; 44:15-24. [PMID: 38305333 DOI: 10.1615/critrevimmunol.2023050273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Sepsis is a life-threatening organ dysfunction due to dysregulated host response to infection, accompanied by a high rate of mortality worldwide. During sepsis progression, toll-like receptors (TLRs) play essential roles in the aberrant inflammatory response that contributes to sepsis-related mortality. Here, we demonstrated a critical role of TLR9 in the progression of sepsis. A septic mouse model was established by cecal ligation and puncture (CLP), then administered with lentivirus encoding si-TLR9/LY294002. TLR9 protein expression and p65 nuclear translocation level/TLR9 protein positive expression/interaction between TLR9 and myeloid differentiation primary response protein 88 (MyD88) in the cecal tissues were examined by Western blot/immunohistochemistry/co-immunoprecipitation assays. Serum levels of pro-inflammatory factors [e.g., interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α)] as well as bacterial contents in the liver/spleen/mesenteric lymph nodes (MLN) were measured by ELISA and bacterial mobility assay. TLR9 expression was augmented in the cecal tissues, TLR9 and MyD88 interaction was enhanced, nuclear p65 protein level was increased, cytoplasmic p65 protein level was decreased, and the nuclear factor kappa B (NF-κB) pathway was activated in CLP-induced septic mice, while TLR9 knockout protected against CLP-induced sepsis via the MyD88/NF-κB pathway inactivation. Briefly, TLR9 inhibition-mediated protection against CLP-induced sepsis was associated with a reduction in pro-inflammatory cytokine release and a promotion of bacterial clearance via a mechanism involving the MyD88/NF-κB pathway inactivation.
Collapse
Affiliation(s)
- Lili Li
- Department of Clinical Laboratory, Fujian Provincial Geriatric Hospital, Fuzhou, Fujian, China
| | - Lili Jiang
- Department of Clinical Laboratory, Fujian Provincial Geriatric Hospital, Fuzhou, Fujian, China
| | - Shuzhu Mao
- Department of Clinical Laboratory, Fujian Provincial Geriatric Hospital, Fuzhou, Fujian, China
| | - Jiajian Ye
- Department of Clinical Laboratory, Fujian Provincial Geriatric Hospital, Fuzhou, Fujian, China
| |
Collapse
|
7
|
Tang W, Xia Y, Deng J, Xu H, Tang Y, Xiao X, Wu L, Song G, Qin J, Wang Y. Anti-inflammatory Effect of Low-Intensity Ultrasound in Septic Rats. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1602-1610. [PMID: 37105771 DOI: 10.1016/j.ultrasmedbio.2023.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/17/2023] [Accepted: 03/07/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVE Sepsis is a severe systemic inflammatory response caused by infection. Here, the spleen region of Sprague-Dawley (SD) rats with sepsis was irradiated with low-intensity ultrasound (LIUS) to explore the regulation of inflammation and its mechanism by LIUS. METHODS In this study, 30 rats used for survival analysis were randomly divided into the sham-operated group (Sham, n = 10), the group in which sepsis was induced by cecal ligation and puncture (CLP, n = 10) and the group treated with LIUS immediately after CLP (LIUS, n = 10). The other 120 rats were randomly divided into the aforementioned three groups for detection at each time point. The parameters used in the LIUS group were 200 mW/cm2, 0.37 MHz, 20% duty cycle and 20 min, and no ultrasonic energy was produced in the Sham and CLP groups. Seven-day survival rate, histopathology and expression of inflammatory factors and proteins were evaluated in the three groups. RESULTS LIUS was able to improve the survival rate of septic SD rats (p < 0.05), significantly inhibit the expression of tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), interleukin 6 (IL-6) and nuclear factor-κB p65 (NF-κB p65) (p < 0.05) and restore the ultrastructure of the spleen. CONCLUSION Our study determined that LIUS can relieve spleen damage and alleviate severe cytokine storm to improve survival outcomes in septic SD rats, and its mechanism may be related to the inhibition of the NF-κB signaling pathway by downregulation of IL-1β.
Collapse
Affiliation(s)
- Wentao Tang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Yi Xia
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Juan Deng
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Haopeng Xu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Yilin Tang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Xinfang Xiao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Liu Wu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Guolin Song
- Department of Emergency, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guizhou, China.
| | - Juan Qin
- Department of Obstetrics and Gynecology, Guiyang Maternal and Child Health Care Hospital, Guizhou Medical University, Guizhou, China.
| | - Yan Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
8
|
Hazrati A, Mirsanei Z, Heidari N, Malekpour K, Rahmani-Kukia N, Abbasi A, Soudi S. The potential application of encapsulated exosomes: A new approach to increase exosomes therapeutic efficacy. Biomed Pharmacother 2023; 162:114615. [PMID: 37011484 DOI: 10.1016/j.biopha.2023.114615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
Cell therapy is one of the methods that have shown promising results in treating diseases in recent decades. However, the use of different types of cells comes with limitations. The application of immune cells in cell therapy can lead to cytokine storms and inappropriate responses to self-antigens. Also, the use of stem cells has the potential to create tumors. Also, cells may not migrate to the injury site after intravenous injection. Therefore, using exosomes from different cells as therapeutic candidates were proposed. Due to their small size and favorable characteristics, such as biocompatibility and immunocompatibility, the easy storage and isolation, exosomes have attracted much attention. They are used in treating many diseases, including cardiovascular diseases, orthopedic diseases, autoimmune diseases, and cancer. However, the results of various studies have shown that the therapeutic efficiency of exosomes (Exo) can be increased by loading different drugs and microRNAs inside them (encapsulated exosomes). Therefore, analyzing studies investigating encapsulated exosomes' therapeutic ability is critical. In this study, we have examined the studies related to the use of encapsulated exosomes in treating diseases such as cancer and infectious diseases and their use in regenerative medicine. Compared to intact exosomes, the results show that the application of encapsulated exosomes has a higher therapeutic ability. Therefore it is suggested to use this method depending on the treatment type to increase the treatment's efficiency.
Collapse
|
9
|
Khosrojerdi A, Soudi S, Hosseini AZ, Khaligh SG, Hashemi SM. The combination of mesenchymal stem cell- and hepatocyte-derived exosomes, along with imipenem, ameliorates inflammatory responses and liver damage in a sepsis mouse model. Life Sci 2023; 326:121813. [PMID: 37257578 DOI: 10.1016/j.lfs.2023.121813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
Aim Sepsis is a medical emergency with no definitive treatment. Animal experiments have confirmed the therapeutic characteristics of exosomes in reducing inflammation and tissue damage. The study investigates the effect of MSC and hepatocyte-derived exosomes along with imipenem in controlling systemic and local (liver) inflammation in a mouse model of sepsis. MAIN METHODS To induce sepsis in C57BL/6 mice, the Cecal Ligation and Puncture (CLP) model was used. The mice were given various treatments, including imipenem, MSC-derived exosomes, hepatocyte-derived exosomes, and a mixture of exosomes. Blood and liver samples were collected and analyzed for cell blood count, liver enzymes, NO levels, cytokine concentrations, and bacterial presence. The percentages of TCD3 + CD4+/CD8+ and Treg in the spleen and mesenteric lymph nodes were also assessed using flow cytometry. The pathological changes were assessed in the liver, lung, and heart tissues. In addition, the cytokine content of exosomes was measured by ELISA. KEY FINDINGS Our results demonstrated that MSC-derived exosomes+imipenem could control systemic and local inflammation and increase the TCD4+ and Treg populations. Hepatocyte-derived exosomes+imipenem reduced inflammation in the liver and increased the TCD8+ and Treg populations. The mixture of exosomes+imipenem had the best function in reducing inflammation, maintaining all T lymphocyte populations, reducing liver damage, and ultimately increasing the survival rate. SIGNIFICANCE The mixture of exosomes derived from MSCs and hepatocytes, along with imipenem, in the inflammatory phase of sepsis could be a promising therapeutic strategy in sepsis treatment.
Collapse
Affiliation(s)
- Arezou Khosrojerdi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Ahmad Zavaran Hosseini
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sahar Ghaffari Khaligh
- Department of Pathology, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Hazrati A, Malekpour K, Soudi S, Hashemi SM. Mesenchymal Stromal/Stem Cells and Their Extracellular Vesicles Application in Acute and Chronic Inflammatory Liver Diseases: Emphasizing on the Anti-Fibrotic and Immunomodulatory Mechanisms. Front Immunol 2022; 13:865888. [PMID: 35464407 PMCID: PMC9021384 DOI: 10.3389/fimmu.2022.865888] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
Abstract
Various factors, including viral and bacterial infections, autoimmune responses, diabetes, drugs, alcohol abuse, and fat deposition, can damage liver tissue and impair its function. These factors affect the liver tissue and lead to acute and chronic liver damage, and if left untreated, can eventually lead to cirrhosis, fibrosis, and liver carcinoma. The main treatment for these disorders is liver transplantation. Still, given the few tissue donors, problems with tissue rejection, immunosuppression caused by medications taken while receiving tissue, and the high cost of transplantation, liver transplantation have been limited. Therefore, finding alternative treatments that do not have the mentioned problems is significant. Cell therapy is one of the treatments that has received a lot of attention today. Hepatocytes and mesenchymal stromal/stem cells (MSCs) are used in many patients to treat liver-related diseases. In the meantime, the use of mesenchymal stem cells has been studied more than other cells due to their favourable characteristics and has reduced the need for liver transplantation. These cells increase the regeneration and repair of liver tissue through various mechanisms, including migration to the site of liver injury, differentiation into liver cells, production of extracellular vesicles (EVs), secretion of various growth factors, and regulation of the immune system. Notably, cell therapy is not entirely excellent and has problems such as cell rejection, undesirable differentiation, accumulation in unwanted locations, and potential tumorigenesis. Therefore, the application of MSCs derived EVs, including exosomes, can help treat liver disease and prevent its progression. Exosomes can prevent apoptosis and induce proliferation by transferring different cargos to the target cell. In addition, these vesicles have been shown to transport hepatocyte growth factor (HGF) and can promote the hepatocytes'(one of the most important cells in the liver parenchyma) growths.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|