1
|
Reive BS, Lau V, Sánchez-Lafuente CL, Henri-Bhargava A, Kalynchuk LE, Tremblay MÈ, Caruncho HJ. The Inflammation-Induced Dysregulation of Reelin Homeostasis Hypothesis of Alzheimer's Disease. J Alzheimers Dis 2024; 100:1099-1119. [PMID: 38995785 PMCID: PMC11380287 DOI: 10.3233/jad-240088] [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] [Indexed: 07/14/2024]
Abstract
Alzheimer's disease (AD) accounts for most dementia cases, but we lack a complete understanding of the mechanisms responsible for the core pathology associated with the disease (e.g., amyloid plaque and neurofibrillary tangles). Inflammation has been identified as a key contributor of AD pathology, with recent evidence pointing towards Reelin dysregulation as being associated with inflammation. Here we describe Reelin signaling and outline existing research involving Reelin signaling in AD and inflammation. Research is described pertaining to the inflammatory and immunological functions of Reelin before we propose a mechanism through which inflammation renders Reelin susceptible to dysregulation resulting in the induction and exacerbation of AD pathology. Based on this hypothesis, it is predicted that disorders of both inflammation (including peripheral inflammation and neuroinflammation) and Reelin dysregulation (including disorders associated with upregulated Reelin expression and disorders of Reelin downregulation) have elevated risk of developing AD. We conclude with a description of AD risk in various disorders involving Reelin dysregulation and inflammation.
Collapse
Affiliation(s)
- Brady S Reive
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Victor Lau
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | - Alexandre Henri-Bhargava
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Vancouver Island Health Authority, Victoria, BC, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Mental Health Research Cluster, University of Victoria, Victoria, BC, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Mental Health Research Cluster, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
2
|
Willmer AR, Diaz-Espinosa J, Zhou A, Stringer KA, Rosania GR. Distinguishing the Concentration- vs. Bioaccumulation-Dependent Immunological and Metabolic Effects of Clofazimine. Pharmaceutics 2023; 15:2350. [PMID: 37765318 PMCID: PMC10537750 DOI: 10.3390/pharmaceutics15092350] [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: 08/19/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
The antimycobacterial drug clofazimine (CFZ) is used as a single agent at high doses, to suppress the exaggerated inflammation associated with leprosy. Paradoxically, increasing doses of CFZ leads to bioaccumulation of CFZ in the spleen and other organs under physiologically relevant dosing regimens, without accompanying dose-dependent elevation in the concentrations of the circulating drug in the blood. In long-term oral dosing regimens, CFZ induces immunological and metabolic changes resulting in splenomegaly, while the mass of other organs decreases or remains unchanged. As an organ that extensively sequesters CFZ as insoluble drug precipitates, the spleen likely influences drug-induced inflammatory signaling. To probe the role of systemic drug concentrations vs. drug bioaccumulation in the spleen, healthy mice were treated with six different dosing regimens. A subgroup of these mice underwent surgical splenectomies prior to drug treatment to assess the bioaccumulation-dependent changes in immune system signaling and immune-system-mediated drug distribution. Under increasing drug loading, the spleen was observed to grow up to six times in size, sequestering over 10% of the total drug load. Interestingly, when the spleen was removed prior to CFZ administration, drug distribution in the rest of the organism was unaffected. However, there were profound cytokine elevations in the serum of asplenic CFZ-treated mice, indicating that the spleen is primarily involved in suppressing the inflammatory signaling mechanisms that are upregulated during CFZ bioaccumulation. Thus, beyond its role in drug sequestration, the spleen actively modulates the systemic effect of CFZ on the immune system, without impacting its blood concentrations or distribution to the rest of the organism.
Collapse
Affiliation(s)
- Andrew R Willmer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jennifer Diaz-Espinosa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Austin Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathleen A Stringer
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gus R Rosania
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
3
|
Li JY, Yao RQ, Xie MY, Zhou QY, Zhao PY, Tian YP, Yao YM. Publication trends of research on sepsis and programmed cell death during 2002–2022: A 20-year bibliometric analysis. Front Cell Infect Microbiol 2022; 12:999569. [PMID: 36211966 PMCID: PMC9537822 DOI: 10.3389/fcimb.2022.999569] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background Sepsis is considered an intractable dysfunction that results from the disordered host immune response to uncontrolled infection. Even though the precise mechanism of sepsis remains unclear, scientific advances have highlighted the key role of various programmed cell death processes in the pathophysiology of sepsis. The current study aims to explore the worldwide research trend on programmed cell death in the setting of sepsis and assesses the achievements of publications from various countries, institutions, journals, and authors globally. Material and methods Associated publications during 2002–2022 with the topical subject of sepsis and programmed cell death were extracted from the Web of Science. VOSviewer was utilized to evaluate and map the published trend in the relevant fields. Results All 2,037 relevant manuscripts with a total citation of 71,575 times were screened out by the end of 1 January 2022. China accounted for the largest number of publications (45.07%) and was accompanied by corporate citations (11,037) and H-index (48), which ranked second globally. The United States has been ranked first place with the highest citations (30,775) and H-index (88), despite a low publication number (29.95%), which was subsequent to China. The journal Shock accounted for the largest number of publications in this area. R. S. Hotchkiss, affiliated with Washington University, was considered to have published the most papers in the relevant fields (57) and achieved the highest citation frequencies (9,523). The primary keywords on the topic of programmed cell death in sepsis remarkably focused on “inflammation” “immunosuppression”, and “oxidative stress”, which were recognized as the core mechanisms of sepsis, eventually attributing to programmed cell death. The involved research on programmed cell death induced by immune dysregulation of sepsis was undoubtedly the hotspot in the pertinent areas. Conclusions The United States has been academically outstanding in sepsis-related research. There appears to be an incompatible performance between publications and quantity with China. Frontier advances may be consulted in the journal Shock. The leading-edge research on the scope of programmed cell death in sepsis should preferably focus on immune dissonance-related studies in the future.
Collapse
Affiliation(s)
- Jing-yan Li
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ren-qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Min-yue Xie
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qi-yuan Zhou
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Peng-yue Zhao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Ying-ping Tian
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Ying-ping Tian, ; Yong-ming Yao,
| | - Yong-ming Yao
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Ying-ping Tian, ; Yong-ming Yao,
| |
Collapse
|
4
|
Qiao J, Cui L. Multi-Omics Techniques Make it Possible to Analyze Sepsis-Associated Acute Kidney Injury Comprehensively. Front Immunol 2022; 13:905601. [PMID: 35874763 PMCID: PMC9300837 DOI: 10.3389/fimmu.2022.905601] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/10/2022] [Indexed: 12/29/2022] Open
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is a common complication in critically ill patients with high morbidity and mortality. SA-AKI varies considerably in disease presentation, progression, and response to treatment, highlighting the heterogeneity of the underlying biological mechanisms. In this review, we briefly describe the pathophysiology of SA-AKI, biomarkers, reference databases, and available omics techniques. Advances in omics technology allow for comprehensive analysis of SA-AKI, and the integration of multiple omics provides an opportunity to understand the information flow behind the disease. These approaches will drive a shift in current paradigms for the prevention, diagnosis, and staging and provide the renal community with significant advances in precision medicine in SA-AKI analysis.
Collapse
Affiliation(s)
- Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, China
- *Correspondence: Liyan Cui,
| |
Collapse
|
5
|
Gao YL, Liu YC, Zhang X, Shou ST, Chai YF. Insight Into Regulatory T Cells in Sepsis-Associated Encephalopathy. Front Neurol 2022; 13:830784. [PMID: 35370925 PMCID: PMC8965708 DOI: 10.3389/fneur.2022.830784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/18/2022] [Indexed: 01/09/2023] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a diffuse central nervous system (CNS) dysfunction during sepsis, and is associated with increased mortality and poor outcomes in septic patients. Despite the high incidence and clinical relevance, the exact mechanisms driving SAE pathogenesis are not yet fully understood, and no specific therapeutic strategies are available. Regulatory T cells (Tregs) have a role in SAE pathogenesis, thought to be related with alleviation of sepsis-induced hyper-inflammation and immune responses, promotion of T helper (Th) 2 cells functional shift, neuroinflammation resolution, improvement of the blood-brain barrier (BBB) function, among others. Moreover, in a clinical point of view, these cells have the potential value of improving neurological and psychiatric/mental symptoms in SAE patients. This review aims to provide a general overview of SAE from its initial clinical presentation to long-term cognitive impairment and summarizes the main features of its pathogenesis. Additionally, a detailed overview on the main mechanisms by which Tregs may impact SAE pathogenesis is given. Finally, and considering that Tregs may be a novel target for immunomodulatory intervention in SAE, different therapeutic options, aiming to boost peripheral and brain infiltration of Tregs, are discussed.
Collapse
Affiliation(s)
- Yu-lei Gao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Yu-lei Gao
| | - Yan-cun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiang Zhang
- Department of Emergency Medicine, Rizhao People's Hospital of Shandong Province, Rizhao, China
| | - Song-tao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan-fen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Yan-fen Chai
| |
Collapse
|
6
|
Gao YL, Yao Y, Zhang X, Chen F, Meng XL, Chen XS, Wang CL, Liu YC, Tian X, Shou ST, Chai YF. Regulatory T Cells: Angels or Demons in the Pathophysiology of Sepsis? Front Immunol 2022; 13:829210. [PMID: 35281010 PMCID: PMC8914284 DOI: 10.3389/fimmu.2022.829210] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a syndrome characterized by life-threatening organ dysfunction caused by the dysregulated host response to an infection. Sepsis, especially septic shock and multiple organ dysfunction is a medical emergency associated with high morbidity, high mortality, and prolonged after-effects. Over the past 20 years, regulatory T cells (Tregs) have been a key topic of focus in all stages of sepsis research. Tregs play a controversial role in sepsis based on their heterogeneous characteristics, complex organ/tissue-specific patterns in the host, the multi-dimensional heterogeneous syndrome of sepsis, the different types of pathogenic microbiology, and even different types of laboratory research models and clinical research methods. In the context of sepsis, Tregs may be considered both angels and demons. We propose that the symptoms and signs of sepsis can be attenuated by regulating Tregs. This review summarizes the controversial roles and Treg checkpoints in sepsis.
Collapse
Affiliation(s)
- Yu-lei Gao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Yan-fen Chai, ; Yu-lei Gao,
| | - Ying Yao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiang Zhang
- Department of Emergency Medicine, Rizhao People’s Hospital of Shandong Province, Rizhao, China
| | - Fang Chen
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiang-long Meng
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin-sen Chen
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao-lan Wang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan-cun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Tian
- Department of Medical Research, Beijing Qiansong Technology Development Company, Beijing, China
| | - Song-tao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan-fen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Yan-fen Chai, ; Yu-lei Gao,
| |
Collapse
|
7
|
Chen H, Huang N, Li J, Sun J, Shi L, Zhang C, Zhao Y, Kong G, Li Z. Immune suppression reversal of the spleen: a promising strategy for improving the survival rate of sepsis in rats. Am J Transl Res 2021; 13:9005-9014. [PMID: 34540012 PMCID: PMC8430070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 06/05/2021] [Indexed: 06/13/2023]
Abstract
Evidence suggests that immune dysfunction exerts a central role in the morbidity and mortality of sepsis. As the spleen is the largest lymphatic tissue in the body, its influence on immune regulation during sepsis should be explored. In this study, we analysed the immune alterations of the spleen of septic rats and the effects of splenectomy at 6 h, 12 h, and 24 h following caecal ligation and puncture (CLP). Results showed declines in CD4+ T cells and elevations in lymphocyte apoptosis, the percentage of Treg cells, and inflammatory cytokine levels (TNF-α, IL-6, and IL-10) in the spleens of CLP-induced septic rats. Moreover, splenectomy improved the survival of septic rats and bacterial clearance from peripheral blood. CLP-induced apoptosis of lymphocytes and the decreased CD4+ T cell percentage in the peripheral blood could be reversed in splenectomy-treated rats. Splenectomy greatly decreased the number of white blood cells, lymphocytes, monocytes, neutrophils, and serum concentration of TNF-α and IL-10 after CLP. Moreover, splenectomy alleviated pathologic damage to the liver and lungs and weakened expression of CD163. These novel findings demonstrate that immune disorders of the spleen are important pathogenic factors during the course of severe sepsis. Splenectomy could alleviate apoptosis and reduction of lymphocytes induced by sepsis, and lower the level of inflammation in the body. Reversing the immune suppression of the spleen may be a novel strategy to improve sepsis survival.
Collapse
Affiliation(s)
- Haiyan Chen
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
- Core Research Laboratory, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
| | - Na Huang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
- Core Research Laboratory, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
| | - Jun Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
| | - Jin Sun
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
| | - Luyi Shi
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
| | - Chen Zhang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
| | - Yang Zhao
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
| | - Guangyao Kong
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
| | - Zongfang Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710004, Shaanxi, China
| |
Collapse
|
8
|
Chen H, Huang N, Tian H, Li J, Li B, Sun J, Zhang S, Zhang C, Zhao Y, Kong G, Li Z. Splenectomy provides protective effects against CLP-induced sepsis by reducing TRegs and PD-1/PD-L1 expression. Int J Biochem Cell Biol 2021; 136:105970. [PMID: 33774183 DOI: 10.1016/j.biocel.2021.105970] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 11/24/2022]
Abstract
The role of the spleen in sepsis is still controversial. Therefore, we investigated the effect of the spleen on sepsis-induced immune dysfunction in C57BL/6 mice subjected to caecal ligation and puncture (CLP). Changes in different immune cells and apoptotic cells in the spleen and peripheral blood were observed 4, 24 and 48 h after CLP. Then, we determined that 48 h following CLP was the most significant period of immunosuppression. Next, we divided the mice into four groups: control, CLP, CLP + spx (splenectomy 48 h after CLP) and spx + CLP (splenectomy surgery two weeks before CLP). Compared with the CLP mice, the CLP + spx and spx + CLP mice had improved survival rates and organ injuries, increased expression of inflammatory factors, a decreased proportion of regulatory T cells (Tregs), and reduced expression of the genes involved in the programmed cell death 1 and its ligand 1 (PD1-PDL1) pathway in immune cells and T-cell immunoglobulin-mucin domain 3 (Tim 3) and Galectin9 in the liver and lungs after 72 h in late-phase sepsis. In addition, the expression of PD-1 was significantly reduced in T cells in spx + CLP mice, and the expression of PD-L1 in myeloid-derived suppressor cells (MDSCs) was reduced in the CLP + spx group, especially in macrophages. These findings suggested that splenectomy could protect septic mice from exhaustion of immune cells by reducing the proliferation of Treg cells and expression of the PD-1/PD-L1 axis in immune cells during the immunosuppressive stage of sepsis. Splenectomy could also reduce liver and lung injuries possibly via the Tim 3 and/or Galectin-9 axis. The spleen is an important regulator of the occurrence and development of sepsis, which provides a new perspective to improve the prognosis of sepsis by regulating the spleen.
Collapse
Affiliation(s)
- Haiyan Chen
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China; Core Research Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Na Huang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China; Core Research Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Hongwei Tian
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jun Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Baohua Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China; Core Research Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jin Sun
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Shaoying Zhang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Chen Zhang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Yang Zhao
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Guangyao Kong
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China; Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Zongfang Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China; Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China.
| |
Collapse
|
9
|
Jergović M, Thompson HL, Bradshaw CM, Sonar SA, Ashgar A, Mohty N, Joseph B, Fain MJ, Cleveland K, Schnellman RG, Nikolich-Žugich J. IL-6 can singlehandedly drive many features of frailty in mice. GeroScience 2021; 43:539-549. [PMID: 33629207 PMCID: PMC8110675 DOI: 10.1007/s11357-021-00343-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 11/30/2022] Open
Abstract
Frailty is a geriatric syndrome characterized by age-related declines in function and reserve resulting in increased vulnerability to stressors. The most consistent laboratory finding in frail subjects is elevation of serum IL-6, but it is unclear whether IL-6 is a causal driver of frailty. Here, we characterize a new mouse model of inducible IL-6 expression (IL-6TET-ON/+ mice) following administration of doxycycline (Dox) in food. In this model, IL-6 induction was Dox dose-dependent. The Dox dose that increased IL-6 levels to those observed in frail old mice directly led to an increase in frailty index, decrease in grip strength, and disrupted muscle mitochondrial homeostasis. Littermate mice lacking the knock-in construct failed to exhibit frailty after Dox feeding. Both naturally old mice and young Dox-induced IL-6TET-ON/+ mice exhibited increased IL-6 levels in sera and spleen homogenates but not in other tissues. Moreover, Dox-induced IL-6TET-ON/+ mice exhibited selective elevation in IL-6 but not in other cytokines. Finally, bone marrow chimera and splenectomy experiments demonstrated that non-hematopoietic cells are the key source of IL-6 in our model. We conclude that elevated IL-6 serum levels directly drive age-related frailty, possibly via mitochondrial mechanisms.
Collapse
Affiliation(s)
- Mladen Jergović
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ USA ,University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, P.O.Box. 249221, 1501 N. Campbell Ave., Tucson, AZ 85724 USA
| | - Heather L. Thompson
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ USA ,University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, P.O.Box. 249221, 1501 N. Campbell Ave., Tucson, AZ 85724 USA ,Present Address: Ventana-Roche Medical Systems, Oro Valley, AZ USA
| | - Christine M. Bradshaw
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ USA ,University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, P.O.Box. 249221, 1501 N. Campbell Ave., Tucson, AZ 85724 USA
| | - Sandip Ashok Sonar
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ USA ,University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, P.O.Box. 249221, 1501 N. Campbell Ave., Tucson, AZ 85724 USA
| | - Arveen Ashgar
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ USA ,University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, P.O.Box. 249221, 1501 N. Campbell Ave., Tucson, AZ 85724 USA
| | - Niels Mohty
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ USA ,University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, P.O.Box. 249221, 1501 N. Campbell Ave., Tucson, AZ 85724 USA
| | - Bellal Joseph
- Division of Trauma Surgery, Department of Surgery, University of Arizona College of Medicine-Tucson, Tucson, AZ USA
| | - Mindy J. Fain
- University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, P.O.Box. 249221, 1501 N. Campbell Ave., Tucson, AZ 85724 USA ,Division of Geriatrics, General Internal Medicine and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ USA
| | - Kristan Cleveland
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ USA
| | - Rick G. Schnellman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ USA ,Southern Arizona Veterans Affairs Health Care System, Tucson, AZ USA ,Southwest Environmental Health Science Center, University of Arizona, Tucson, AZ USA
| | - Janko Nikolich-Žugich
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA. .,University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, P.O.Box. 249221, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA.
| |
Collapse
|
10
|
Piao J, Park JS, Hwang DY, Hong HS, Son Y. Substance P blocks β-aminopropionitrile-induced aortic injury through modulation of M2 monocyte-skewed monocytopoiesis. Transl Res 2021; 228:76-93. [PMID: 32835906 DOI: 10.1016/j.trsl.2020.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/27/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
Aortic injuries, including aortic aneurysms and dissections, are fatal vascular diseases with distinct histopathological features in the aortic tissue such as inflammation-induced endothelial dysfunction, infiltration of immune cells, and breakdown of the extracellular matrix. Few treatments are available for treating aortic aneurysms and dissections; thus, basic and clinical studies worldwide have been attempted to inhibit disease progression. Substance P (SP) exerts anti-inflammatory effects and promotes restoration of the damaged endothelium, leading to vasculature protection and facilitation of tissue repair. This study was conducted to explore the protective effects of systemically injected SP on thoracic aortic injury (TAI). A TAI animal model was induced by orally administering β-aminopropionitrile to rats for 6 weeks. β-aminopropionitrile blocked crosslinking ECM in aorta to cause structural alteration with inflammation within 1 week and then, induced aortic dissection within 4 weeks of initiating treatment, leading to mortality within 6 weeks. Treatment of TAI rats with SP-induced anti-inflammatory responses systemically and locally, possibly by enriching anti-inflammatory M2 monocytes in the spleen and peripheral blood at early phase of aortic injury due to β-aminopropionitrile. SP-induced immune suppression finally prevented the development of aortic dissection by limiting inflammation-mediated aortic destruction. Taken together, these results suggest that SP treatment can block aortic injury by controlling the immune-cell profile and suppressing proinflammatory responses during the initial stage of vascular disease progression.
Collapse
Affiliation(s)
- Jiyuan Piao
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yong In, South Korea
| | - Jeong Seop Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Dae Yeon Hwang
- East-West Medical Research Institute, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, South Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, South Korea; East-West Medical Research Institute, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, South Korea.
| | - Youngsook Son
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yong In, South Korea.
| |
Collapse
|
11
|
A key role of gut microbiota-vagus nerve/spleen axis in sleep deprivation-mediated aggravation of systemic inflammation after LPS administration. Life Sci 2020; 265:118736. [PMID: 33176177 DOI: 10.1016/j.lfs.2020.118736] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/26/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022]
Abstract
AIMS Sleep deprivation (SD) correlates with exacerbated systemic inflammation after sepsis. However, the underlying mechanisms remain unclear. This study aimed to evaluate the roles and mechanisms of SD in inflammatory organ injury after lipopolysaccharide (LPS) administration. MAIN METHODS Mice were intraperitoneally injected with LPS followed by 3 consecutive days of SD. The pseudo germ-free (PGF) mice received fecal microbiota transplant by being gavaged with supernatant from fecal suspension of septic mice with or without SD. The subdiaphragmatic vagotomy (SDV) or splenectomy was performed 14 days prior to LPS injection or antibiotics administration. KEY FINDINGS Post-septic SD increased the plasma levels of interleukin (IL)-6 and tumor necrosis factor-α (TNF-α), reduced IL-10 plasma level, increased spleen weight, and promoted inflammatory injury of the lung, liver and kidney. The relative abundance of Proteobacteria and its subgroups were increased after post-septic SD. PGF mice transplanted with fecal bacteria from septic mice subjected to SD developed splenomegaly, systemic inflammation, organ inflammation and damage as their donors did. Intriguingly, SDV abolished the aggravated effects of SD on splenomegaly and inflammatory organ injury in septic mice received SD or in PGF mice transplanted with fecal bacteria from septic mice subjected to SD. Furthermore, splenectomy also abrogated the increase in IL-6 and TNF-α plasma levels and the decrease in IL-10 plasma level in PGF mice transplanted with fecal bacteria from septic mice subjected to SD. SIGNIFICANCE Gut microbiota-vagus nerve axis and gut microbiota-spleen axis play key roles in modulating systemic inflammation induced by SD after LPS administration.
Collapse
|
12
|
Cheng Z, Abrams ST, Toh J, Wang SS, Wang Z, Yu Q, Yu W, Toh CH, Wang G. The Critical Roles and Mechanisms of Immune Cell Death in Sepsis. Front Immunol 2020; 11:1918. [PMID: 32983116 PMCID: PMC7477075 DOI: 10.3389/fimmu.2020.01918] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 07/16/2020] [Indexed: 12/23/2022] Open
Abstract
Sepsis was first described by the ancient Greek physicians over 2000 years ago. The pathophysiology of the disease, however, is still not fully understood and hence the mortality rate is still unacceptably high due to lack of specific therapies. In the last decade, great progress has been made by shifting the focus of research from systemic inflammatory response syndrome (SIRS) to multiple organ dysfunction syndrome (MODS). Sepsis has been re-defined as infection-induced MODS in 2016. How infection leads to MODS is not clear, but what mediates MODS becomes the major topic in understanding the molecular mechanisms and developing specific therapies. Recently, the mechanism of infection-induced extensive immune cell death which releases a large quantity of damage-associated molecular patterns (DAMPs) and their roles in the development of MODS as well as immunosuppression during sepsis have attracted much attention. Growing evidence supports the hypothesis that DAMPs, including high-mobility group box 1 protein (HMGB1), cell-free DNA (cfDNA) and histones as well as neutrophil extracellular traps (NETs), may directly or indirectly contribute significantly to the development of MODS. Here, we provide an overview of the mechanisms and consequences of infection-induced extensive immune cell death during the development of sepsis. We also propose a pivotal pathway from a local infection to eventual sepsis and a potential combined therapeutic strategy for targeting sepsis.
Collapse
Affiliation(s)
- Zhenxing Cheng
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Medical School, Southeast University, Nanjing, China
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Julien Toh
- Wirral University Teaching Hospitals NHS Foundation Trust, Wirral, United Kingdom
| | | | - Zhi Wang
- Medical School, Southeast University, Nanjing, China
| | - Qian Yu
- Medical School, Southeast University, Nanjing, China
| | - Weiping Yu
- Medical School, Southeast University, Nanjing, China
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Medical School, Southeast University, Nanjing, China
| |
Collapse
|
13
|
Li W, Li L, Li W, Chopp M, Venkat P, Zacharek A, Chen Z, Landschoot-Ward J, Chen J. Spleen associated immune-response mediates brain-heart interaction after intracerebral hemorrhage. Exp Neurol 2020; 327:113209. [PMID: 31987832 DOI: 10.1016/j.expneurol.2020.113209] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/06/2020] [Accepted: 01/24/2020] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND PURPOSE Intracerebral hemorrhage (ICH) patients frequently encounter cardiovascular complications which may contribute to increased mortality and poor long term outcome. ICH induces systemic oxidative stress and activates peripheral immune responses which are involved in the pathological cascade leading to cardiac dysfunction and heart failure after ICH. We have previously reported that ICH induces progressive cardiac dysfunction in mice without primary cardiac diseases. In this study, we have investigated the role of immune response in mediating cardiac dysfunction post ICH in mice. METHODS Adult male C57BL/6 J mice were randomly assigned to the following groups (n = 8/group): 1) sham control; 2) ICH; 3) splenectomy with ICH (ICH + Spx); 4) splenectomy alone (Spx). Echocardiography was performed at 7 and 28 days after ICH. A battery of neurological and cognitive tests were performed. Flow cytometry, western blot and immunostaining were used to test mechanisms of ICH induced cardiac dysfunction. RESULTS Compared to sham control mice, Spx alone does not induce acute (7 day) or chronic (28 day) cardiac dysfunction. ICH induces significant neurological and cognitive deficits, as well as acute and chronic cardiac dysfunction compared to sham control mice. Mice subjected to ICH + Spx exhibit significantly improved neurological and cognitive function compared to ICH mice. Mice with ICH + Spx also exhibit significantly improved acute and chronic cardiac function compared to ICH mice indicated by increased left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS), decreased cardiac fibrosis, decreased cardiomyocyte hypertrophy, decreased cardiac infiltration of immune cells and decreased expression of inflammatory factor and oxidative stress in the heart. CONCLUSIONS Our study demonstrates that splenectomy attenuates ICH-induced neurological and cognitive impairment as well as ICH-induced cardiac dysfunction in mice. Inflammatory cell infiltration into heart and immune responses mediated by the spleen may contribute to ICH-induce acute and chronic cardiac dysfunction and pathological cardiac remodeling.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Linlin Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wenkui Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Zhili Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA.
| |
Collapse
|
14
|
El-Naseery NI, Mousa HSE, Noreldin AE, El-Far AH, Elewa YHA. Aging-associated immunosenescence via alterations in splenic immune cell populations in rat. Life Sci 2019; 241:117168. [PMID: 31838133 DOI: 10.1016/j.lfs.2019.117168] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 12/16/2022]
Abstract
AIM Immunosenescence is the decline of the host immune system due to aging, resulting in various complications. The splenic lymphoid nodule is the pivotal compartment involved in immunosenescence. In this study, we investigated the important changes in the splenic immune cell populations of aged rats (18-24 months) in comparison with young rats (3-5 months). MATERIALS AND METHODS We, also, studied the effects of aging on the activities of total superoxide dismutase (T-SOD) and malondialdehyde (MDA) levels in spleen of both groups, besides the changes of the splenic architecture. Furthermore, immunohistochemical staining was performed to detect the aging effects in T cells, B cells, macrophages, granulocytes, mast cells, proliferating cells, apoptotic cells, and cells positive for interleukin-1β (IL-1β), interleukin-6 (IL-6), and Toll-like receptor 4 (TLR4). KEY FINDINGS The aged rats had significantly lower spleen/body weight ratios and smaller splenic nodules, indicating a decline in general immunity in them. With aging, T-SOD activities were decreased, while MDA levels were increased, exhibiting that oxidative stress increases in spleens. In addition, the aged group also had significantly fewer T and B cells, macrophages, granulocytes, IL-6 and TLR4 immuno-positive cells, and proliferating cells in the periarterial lymphatic sheaths, marginal zone, and lymphoid follicles compared with the young group. On the other hand, the number of mast cells and apoptotic cells was significantly increased with age. Therefore, we can conclude that cellular immunity and humoral immunity were crumpled with age.
Collapse
Affiliation(s)
- Nesma I El-Naseery
- Histology and Cytology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Hanaa S E Mousa
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Ahmed E Noreldin
- Histology and Cytology Department, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Yaser H A Elewa
- Histology and Cytology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt; Laboratory of Anatomy, Faculty of Veterinary Medicine, Basic Veterinary Sciences, Hokkaido University, Sapporo 060-0818, Japan.
| |
Collapse
|
15
|
Comparison of post-traumatic changes in circulating and bone marrow leukocytes between BALB/c and CD-1 mouse strains. PLoS One 2019; 14:e0222594. [PMID: 31527918 PMCID: PMC6748677 DOI: 10.1371/journal.pone.0222594] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 09/03/2019] [Indexed: 11/19/2022] Open
Abstract
This manuscript emerged from a larger third-party funded project investigating a new poly-trauma model and its influence upon secondary sepsis. The present sub-study compared selected leukocyte subpopulations in the circulation and bone marrow after polytrauma in BALB/c versus CD-1 mice. Animals underwent unilateral femur fracture, splenectomy and hemorrhagic shock. We collected blood and bone marrow for flow cytometry analysis at 24h and 48h post-trauma. Circulating granulocytes (Ly6G+CD11+) increased in both strains after trauma. Only in BALB/c mice circulating CD8+ T-lymphocytes decreased within 48h by 30%. Regulatory T-cells (Tregs, CD4+CD25+CD127low) increased in both strains by approx. 32%. Circulating Tregs and lymphocytes (CD11b-Ly6G-MHC-2+) were always at least 1.5-fold higher in BALB/c, while the bone marrow MHC-2 expression decreased in CD-1 mice (p<0.05). Overall, immune responses to polytrauma were similar in both strains. Additionally, BALB/c expressed higher level of circulating regulatory T-cells and MHC-2-positive lymphocytes compared to CD-1 mice.
Collapse
|
16
|
Skirecki T, Drechsler S, Hoser G, Jafarmadar M, Siennicka K, Pojda Z, Kawiak J, Osuchowski MF. The Fluctuations of Leukocytes and Circulating Cytokines in Septic Humanized Mice Vary With Outcome. Front Immunol 2019; 10:1427. [PMID: 31297113 PMCID: PMC6607920 DOI: 10.3389/fimmu.2019.01427] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/06/2019] [Indexed: 12/20/2022] Open
Abstract
Sepsis remains a major challenge in translational research given its heterogeneous pathophysiology and the lack of specific therapeutics. The use of humanized mouse chimeras with transplanted human hematopoietic cells may improve the clinical relevance of pre-clinical studies. However, knowledge of the human immuno-inflammatory response during sepsis in humanized mice is scarce; it is unclear how similar or divergent mouse and human-origin immuno-inflammatory responses in sepsis are. In this study, we evaluated the early outcome-dependent immuno-inflammatory response in humanized mice generated in the NSG strain after cecal ligation and puncture (CLP) sepsis. Mice were observed for 32 h post-CLP and were assigned to either predicted-to-die (P-DIE) or predicted-to-survive (P-SUR) groups for retrospective comparisons. Blood samples were collected at baseline, 6 and 24 h, whereas the bone marrow and spleen were collected between 24 and 32 h post-CLP. In comparison to P-SUR, P-DIE humanized mice had a 3-fold higher frequency of human splenic monocytes and their CD80 expression was reduced by 1.3-fold; there was no difference in the HLA-DR expression. Similarly, the expression of CD80 on the bone marrow monocytes from P-DIE mice was decreased by 32% (p < 0.05). Sepsis induced a generalized up-regulation of both human and murine plasma cytokines (TNFα, IL-6, IL-10, IL-8/KC, MCP-1); it was additionally aggravated in P-DIE vs. P-SUR. Human cytokines were strongly overridden by the murine ones (approx. ratio 1:9) but human TNFα was 7-fold higher than mouse TNFα. Interestingly, transplantation of human cells did not influence murine cytokine response in NSG mice, but humanized NSG mice were more susceptible to sepsis in comparison with NSG mice (79 vs. 33% mortality; p < 0.05). In conclusion, our results show that humanized mice reflect selected aspects of human immune responses in sepsis and therefore may be a feasible alternative in preclinical immunotherapy modeling.
Collapse
Affiliation(s)
- Tomasz Skirecki
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Susanne Drechsler
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Research Center, Vienna, Austria
| | - Grazyna Hoser
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Mohammad Jafarmadar
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Research Center, Vienna, Austria
| | - Katarzyna Siennicka
- Department of Regenerative Medicine, Maria Sklodowska-Curie Institute-Oncology Center, Warsaw, Poland
| | - Zygmunt Pojda
- Department of Regenerative Medicine, Maria Sklodowska-Curie Institute-Oncology Center, Warsaw, Poland
| | - Jerzy Kawiak
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Marcin F Osuchowski
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Research Center, Vienna, Austria
| |
Collapse
|