1
|
Briggs GD, Meakes S, King KL, Balogh ZJ. Cell-free mitochondria are detected in high concentrations in the plasma of orthopedic trauma patients. J Trauma Acute Care Surg 2025; 98:760-768. [PMID: 39621451 PMCID: PMC12036778 DOI: 10.1097/ta.0000000000004483] [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: 05/22/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 04/24/2025]
Abstract
BACKGROUND Trauma and surgery can derange inflammatory and hemostasis responses, potentially leading to multiple organ failure. Mitochondrial damage-associated molecular patterns are known to be part of the pathomechanism, but their exact origin remains uncertain. Recently, intact mitochondria were detected in healthy individuals' peripheral blood, which suggested a potential role in inflammation. METHODS In this case-control study, we quantitated cell-free mitochondria in the blood of healthy subjects (n = 4) and trauma patients (n = 25) and assessed their relationship with patient demographics, injury and shock severity, markers of tissue injury, inflammation, and blood transfusions. Blood samples were collected before and after major orthopedic trauma surgery, and cell-free mitochondria were quantified using flow cytometry, targeting the outer mitochondrial membrane protein, TOMM70. Mitotracker Deep Red staining was used to assess mitochondrial membrane potential. RESULTS Trauma patients had significantly more cell-free mitochondria in their plasma compared with healthy controls, with highest counts immediately after surgery. The number of cell-free mitochondria decreased by day 5 postoperatively. Trauma patients exhibited a higher proportion of active cell-free mitochondria compared with healthy controls, especially immediately after surgery, and this proportion correlated with tissue injury markers. Associations were also found with acute thrombocytopenia, Denver multiple organ failure score, and transfusion of fresh frozen plasma and cryoprecipitate. CONCLUSION Our findings indicate that the mere high number of cell-free mitochondria in the circulation of trauma patients is not necessarily pro-inflammatory, but their active status is associated with more severe secondary tissue injury. The natural history of cell-free mitochondria in trauma needs to be characterized, including their potential cause-effect relationship with major postinjury complications. LEVEL OF EVIDENCE Prognostic and Epidemiological; Level III.
Collapse
|
2
|
Haines KL, Li R, Grey S, Kim HE, Gann E, Almond C, Rouse M, Joshi M, Schobel-McHugh S, Agarwal S, Kirk A, Elster E, Fernandez-Moure JS. Exploratory cluster analysis of IL2Ra and associated biomarkers and complications after blunt chest trauma. J Trauma Acute Care Surg 2025:01586154-990000000-00963. [PMID: 40223169 DOI: 10.1097/ta.0000000000004568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
BACKGROUND Rib fractures compromise approximately 40% of all fractures in the United States. Despite their prevalence, the relationship between rib fractures, solid organ injuries, and immune responses remains poorly understood. This exploratory study investigates the immunological profile associated with pulmonary and renal complications in rib fracture patients using data from our Surgical Critical Care Initiative Clinical Data Repository. The aim is to correlate distinct cytokine/chemokine profiles with high-energy rib fracture patterns, such as first rib fracture, and associated complications, potentially providing predictive biomarkers for patient outcomes. METHODS Clinical and demographic data on patients with rib fractures were extracted from Surgical Critical Care Initiative Clinical Data Repository. Patients were categorized based on the presence or absence of complications. A comprehensive panel of 46 inflammation and tissue repair biomarkers was measured using the Meso Scale Discovery platforms. Principal component analysis was used to reduce the dimensionality of the cytokine data. Statistical and machine learning models assessed the association between biomarker patterns, rib fracture localization, and complications. Logistic regression models with high discriminative performance were developed for first rib fractures (high energy transfer), lung injury, and pneumonia. RESULTS Among 150 rib fracture patients, 73 had complications. Cytokine analysis revealed two distinct clusters: Cluster 1, associated with pro-inflammatory responses and tissue repair, and Cluster 2, linked with anti-inflammatory responses, angiogenesis, and immunometabolism. Predictive models demonstrated strong validity (area under the curve, >0.90) and identified key variables such as the cytokine IL2Ra, significantly associated with acute kidney injury, acute lung injury, and pulmonary complications post-rib fractures, particularly first rib fractures. CONCLUSION IL2Ra release is significantly correlated with high-energy transfer injuries like first rib fractures, indicating a bidirectional relationship between these fractures and the immune response. Furthermore, a hierarchical relationship exists among clinical complications, with kidney and lung injuries frequently preceding pneumonia. These findings underscore the potential utility of integrating immunological markers into clinical decision-support frameworks for personalized therapeutic interventions. LEVEL OF EVIDENCE Prognostic; Level III.
Collapse
Affiliation(s)
- Krista L Haines
- From the Division of Trauma and Acute Care Surgery, Department of Surgery (K.L.H., S.A., J.S.F.-M.), Duke University Medical Center, Durham, North Carolina; Department of Surgery (R.L., S.G., H.E.K., E.G., M.R., S.S.-M.), Uniformed Services University of the Health Sciences; Henry M. Jackson Foundation for the Advancement of Military Medicine (R.L., S.G., H.E.K., E.G., M.R., S.S.-M., E.E.), Bethesda, Maryland; Surgical Critical Care Initiative (K.L.H., R.L., S.G., H.E.K., E.G., C.A., M.R., M.J., S.S.-M., S.A., A.K., E.E., J.S.F.-M.), Bethesda, Maryland; Clinical Research Institute (C.A.), Substrate Services Core Research Support (M.J.), and Department of Surgery (A.K.), Duke University, Durham, North Carolina; and Walter Reed National Military Medical Center (E.E.), Bethesda, Maryland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Andrianova NV, Buyan MI, Brezgunova AA, Cherkesova KS, Zorov DB, Plotnikov EY. Hemorrhagic Shock and Mitochondria: Pathophysiology and Therapeutic Approaches. Int J Mol Sci 2025; 26:1843. [PMID: 40076469 PMCID: PMC11898946 DOI: 10.3390/ijms26051843] [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: 12/30/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Severe injuries and some pathologies associated with massive bleeding, such as maternal hemorrhage, gastrointestinal and perioperative bleeding, and rupture of an aneurysm, often lead to major blood loss and the development of hemorrhagic shock. A sharp decrease in circulating blood volume triggers a vicious cycle of vasoconstriction and coagulopathy leading to ischemia of all internal organs and, in severe decompensated states, ischemia of the brain and heart. The basis of tissue damage and dysfunction in hemorrhagic shock is an interruption in the supply of oxygen and substrates for energy production to the cells, making the mitochondria a source and target of oxidative stress and proapoptotic signaling. Based on these mechanisms, different strategies are proposed to treat the multiple organ failure that occurs in shock. The main direction of such treatment is to provide the cells with a sufficient amount of substrates that utilize oxidative phosphorylation at different stages and increase the efficiency of energy production by the mitochondria. These strategies include restoring the efficiency of mitochondrial complexes, for example, by restoring the nicotinamide adenine dinucleotide (NAD) pool. Another direction is approaches to minimize oxidative stress as well as apoptosis, which are primarily dependent on the mitochondria. There are also a number of other methods to reduce mitochondrial dysfunction and improve the quality of the mitochondrial population. In this review, we consider such strategies for the treatment of hemorrhagic shock and show the promise of therapeutic approaches aimed at restoring the bioenergetic functions of the cell and protecting mitochondria.
Collapse
Affiliation(s)
- Nadezda V. Andrianova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
| | - Marina I. Buyan
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Anna A. Brezgunova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
| | - Kseniia S. Cherkesova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- Faculty of Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Dmitry B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia
| |
Collapse
|
4
|
Pan H, Su Q, Hong P, You Y, Zhou L, Zou J, Sun J, Zhong G, Liao J, Zhang H, Tang Z, Hu L. Arsenic-induced mtDNA release promotes inflammatory responses through cGAS-STING signaling in chicken hepatocytes. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 205:106129. [PMID: 39477583 DOI: 10.1016/j.pestbp.2024.106129] [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: 07/25/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 11/07/2024]
Abstract
Arsenic is a toxic element that can cause severe liver damage in humans and animals. Arsenic-based inorganic pesticides, such as lead arsenate, copper arsenate, and calcium arsenate, are widely used for insect control and can eventually affect human health through accumulation in the food chain. However, the relationship between arsenic trioxide (ATO)-induced hepatotoxicity and the cGAS-STING signaling pathway has not been reported. The aim of this study was to investigate the potential role of inflammatory response in ATO-induced hepatotoxicity in chickens. In this study, we found that ATO exposure resulted in mtDNA leakage into the cytoplasm of chicken hepatocytes, which activated the cGAS-STING pathway and significantly increased the cGAS, STING, TBK1, and IRF7 mRNA and protein expression levels. Moreover, type I interferon response was activated. Concurrently, STING triggered the activation of the traditional NF-κB signaling pathway and promoted the expression of pro-inflammatory cytokine genes, including TNF-α, IL-6, and IL-1β. Subsequently, we found that both mtDNA clearance with EtBr and inhibition of the cGAS-STING pathway with H-151 reversed the ATO-induced innate immune and inflammatory responses. In summary, the above findings indicate that chicken hepatocytes can induce innate immune responses and inflammatory responses via mtDNA-cGAS-STING under ATO-exposure conditions, which is of great significance for further studies on the toxicity mechanism of ATO.
Collapse
Affiliation(s)
- Hang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Qian Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Panjing Hong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Yanli You
- College of Life Science, Yantai University, Yantai, 264005, Shandong Province, China.
| | - Limeng Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Junbo Zou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Jingping Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Gaolong Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
5
|
Dobson GP, Letson HL, Morris JL. Revolution in sepsis: a symptoms-based to a systems-based approach? J Biomed Sci 2024; 31:57. [PMID: 38811967 PMCID: PMC11138085 DOI: 10.1186/s12929-024-01043-4] [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: 04/02/2024] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Abstract
Severe infection and sepsis are medical emergencies. High morbidity and mortality are linked to CNS dysfunction, excessive inflammation, immune compromise, coagulopathy and multiple organ dysfunction. Males appear to have a higher risk of mortality than females. Currently, there are few or no effective drug therapies to protect the brain, maintain the blood brain barrier, resolve excessive inflammation and reduce secondary injury in other vital organs. We propose a major reason for lack of progress is a consequence of the treat-as-you-go, single-nodal target approach, rather than a more integrated, systems-based approach. A new revolution is required to better understand how the body responds to an infection, identify new markers to detect its progression and discover new system-acting drugs to treat it. In this review, we present a brief history of sepsis followed by its pathophysiology from a systems' perspective and future opportunities. We argue that targeting the body's early immune-driven CNS-response may improve patient outcomes. If the barrage of PAMPs and DAMPs can be reduced early, we propose the multiple CNS-organ circuits (or axes) will be preserved and secondary injury will be reduced. We have been developing a systems-based, small-volume, fluid therapy comprising adenosine, lidocaine and magnesium (ALM) to treat sepsis and endotoxemia. Our early studies indicate that ALM therapy shifts the CNS from sympathetic to parasympathetic dominance, maintains cardiovascular-endothelial glycocalyx coupling, reduces inflammation, corrects coagulopathy, and maintains tissue O2 supply. Future research will investigate the potential translation to humans.
Collapse
Affiliation(s)
- Geoffrey P Dobson
- Heart, Sepsis and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, 1 James Cook Drive, Townsville, QLD, 4811, Australia.
| | - Hayley L Letson
- Heart, Sepsis and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, 1 James Cook Drive, Townsville, QLD, 4811, Australia
| | - Jodie L Morris
- Heart, Sepsis and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, 1 James Cook Drive, Townsville, QLD, 4811, Australia
| |
Collapse
|
6
|
Torp MK, Stensløkken KO, Vaage J. When Our Best Friend Becomes Our Worst Enemy: The Mitochondrion in Trauma, Surgery, and Critical Illness. J Intensive Care Med 2024:8850666241237715. [PMID: 38505947 DOI: 10.1177/08850666241237715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Common for major surgery, multitrauma, sepsis, and critical illness, is a whole-body inflammation. Tissue injury is able to trigger a generalized inflammatory reaction. Cell death causes release of endogenous structures termed damage associated molecular patterns (DAMPs) that initiate a sterile inflammation. Mitochondria are evolutionary endosymbionts originating from bacteria, containing molecular patterns similar to bacteria. These molecular patterns are termed mitochondrial DAMPs (mDAMPs). Mitochondrial debris released into the extracellular space or into the circulation is immunogenic and damaging secondary to activation of the innate immune system. In the circulation, released mDAMPS are either free or exist in extracellular vesicles, being able to act on every organ and cell in the body. However, the role of mDAMPs in trauma and critical care is not fully clarified. There is a complete lack of knowledge how they may be counteracted in patients. Among mDAMPs are mitochondrial DNA, cardiolipin, N-formyl peptides, cytochrome C, adenosine triphosphate, reactive oxygen species, succinate, and mitochondrial transcription factor A. In this overview, we present the different mDAMPs, their function, release, targets, and inflammatory potential. In light of present knowledge, the role of mDAMPs in the pathophysiology of major surgery and trauma as well as sepsis, and critical care is discussed.
Collapse
Affiliation(s)
- May-Kristin Torp
- Section of Physiology, Department of Molecular Medicine, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
- Department of Research, Østfold Hospital Trust, Grålum, Norway
| | - Kåre-Olav Stensløkken
- Section of Physiology, Department of Molecular Medicine, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
| | - Jarle Vaage
- Section of Physiology, Department of Molecular Medicine, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
- Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
7
|
de Diego C, Lasierra AB, López-Vergara L, Torralba L, Ruiz de Gopegui P, Lahoz R, Abadía C, Godino J, Cebollada A, Jimeno B, Bello C, Tejada A, Bello S. What is the actual relationship between neutrophil extracellular traps and COVID-19 severity? A longitudinal study. Respir Res 2024; 25:48. [PMID: 38243237 PMCID: PMC10797938 DOI: 10.1186/s12931-023-02650-9] [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: 08/02/2023] [Accepted: 12/21/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Neutrophil extracellular traps (NETs) have repeatedly been related to COVID-19 severity and mortality. However, there is no consensus on their quantification, and there are scarce data on their evolution during the disease. We studied circulating NET markers in patients with COVID-19 throughout their hospitalization. METHODS We prospectively included 93 patients (201 blood samples), evaluating the disease severity in 3 evolutionary phases (viral, early, and late inflammation). Of these, 72 had 180 samples in various phases. We also evaluated 55 controls with similar age, sex and comorbidities. We measured 4 NET markers in serum: cfDNA, CitH3, and MPO-DNA and NE-DNA complexes; as well as neutrophil-related cytokines IL-8 and G-CSF. RESULTS The COVID-19 group had higher CitH3 (28.29 vs 20.29 pg/mL, p = 0.022), and cfDNA, MPO-DNA, and NE-DNA (7.87 vs 2.56 ng/mL; 0.80 vs 0.52 and 1.04 vs 0.72, respectively, p < 0.001 for all) than the controls throughout hospitalisation. cfDNA was the only NET marker clearly related to severity, and it remained higher in non-survivors during the 3 phases. Only cfDNA was an independent risk factor for mortality and need for intensive care. Neutrophil count, IL-8, and G-CSF were significantly related to severity. MPO-DNA and NE-DNA showed significant correlations (r: 0.483, p < 0.001), including all 3 phases and across all severity grades, and they only remained significantly higher on days 10-16 of evolution in those who died. Correlations among the other NET markers were lower than expected. CONCLUSIONS The circulating biomarkers of NETs were present in patients with COVID-19 throughout hospitalization. cfDNA was associated with severity and mortality, but the three other markers showed little or no association with these outcomes. Neutrophil activity and neutrophil count were also associated with severity. MPO-DNA and NE-DNA better reflected NET formation. cfDNA appeared to be more associated with overall tissue damage; previous widespread use of this marker could have overestimated the relationship between NETs and severity. Currently, there are limitations to accurate NET markers measurement that make it difficult to assess its true role in COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Cristina de Diego
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel la Católica 1-9, 50009, Zaragoza, Spain
| | | | - Lucía López-Vergara
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel la Católica 1-9, 50009, Zaragoza, Spain
| | - Laura Torralba
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel la Católica 1-9, 50009, Zaragoza, Spain
| | | | - Raquel Lahoz
- Department of Biochemistry. Miguel, Servet University Hospital, Zaragoza, Spain
| | - Claudia Abadía
- Department of Biochemistry. Miguel, Servet University Hospital, Zaragoza, Spain
| | - Javier Godino
- Department of Cytometry and Cell Separation, Aragon Institute of Health Sciences (IACS), Zaragoza, Spain
| | - Alberto Cebollada
- Biocomputing Technical Scientific Service, Aragon Institute of Health Sciences (IACS), Zaragoza, Spain
| | - Beatriz Jimeno
- Department of Cytometry and Cell Separation, Aragon Institute of Health Sciences (IACS), Zaragoza, Spain
| | - Carlota Bello
- Department of Radiology, Hospital Clínico Lozano Blesa, Zaragoza, Spain
| | - Antonio Tejada
- Intensive Care Unit, Miguel Servet University Hospital, Zaragoza, Spain
| | - Salvador Bello
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel la Católica 1-9, 50009, Zaragoza, Spain.
| |
Collapse
|
8
|
Fricchione G. Brain evolution and the meaning of catatonia - An update. Schizophr Res 2024; 263:139-150. [PMID: 36754715 DOI: 10.1016/j.schres.2023.01.026] [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: 10/31/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 02/10/2023]
Abstract
Back in 2004, in a chapter titled "Brain Evolution and the Meaning of Catatonia", a case was made that the syndrome's core meaning is embedded in millions of years of vertebrate brain evolution. (Fricchione, 2004) In this update, advances over the last almost 20 years, in catatonia theory and research in particular, and pertinent neuropsychiatry in general, will be applied to this question of meaning. The approach will rely heavily on a number of thought leaders, including Nicos Tinbergen, Paul MacLean, John Bowlby, M. Marsel Mesulam, Bruce McEwen and Karl Friston. Their guidance will be supplemented with a selected survey of 21sty century neuropsychiatry, neurophysiology, molecular biology, neuroimaging and neurotherapeutics as applied to the catatonic syndrome. In an attempt to address the question of the meaning of the catatonic syndrome in human life, we will employ two conceptual networks representing the intersubjectivity of the quantitative conceptual network of physical terms and the subjectivity of the qualitative conceptual network of mental and spiritual terms. In the process, a common referent providing extensional identity may emerge (Goodman, 1991). The goal of this exercise is to enhance our attunement with the experience of patients suffering with catatonia. A deeper understanding of catatonia's origins in brain evolution and of the challenges of individual epigenetic development in the setting of environmental events coupled with appreciation of what has been described as the most painful mammalian condition, that of separation, has the potential to foster greater efforts on the part of clinicians to diagnose and treat patients who present with catatonia. In addition, in this ancient and extreme tactic, evolved to provide safety from extreme survival threat, one can speculate what is at the core of human fear and the challenge it presents to all of us. And when the biology, psychology and sociology of catatonia are examined, the nature of solutions to the challenge may emerge.
Collapse
Affiliation(s)
- Gregory Fricchione
- Benson-Henry Institute for Mind Body Medicine Division of Psychiatry and Medicine Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Xiao Y, Fang H, Zhu Y, Zhou J, Dai Z, Wang H, Xia Z, Tu Z, Leong KW. Multifunctional Cationic Hyperbranched Polyaminoglycosides that Target Multiple Mediators for Severe Abdominal Trauma Management. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305273. [PMID: 37997512 PMCID: PMC10767409 DOI: 10.1002/advs.202305273] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/12/2023] [Indexed: 11/25/2023]
Abstract
Trauma and its associated complications, including dysregulated inflammatory responses, severe infection, and disseminated intravascular coagulation (DIC), continue to pose lethal threats worldwide. Following injury, cell-free nucleic acids (cfNAs), categorized as damage-associated molecular patterns (DAMPs), are released from dying or dead cells, triggering local and systemic inflammatory responses and coagulation abnormalities that worsen disease progression. Harnessing cfNA scavenging strategies with biomaterials has emerged as a promising approach for treating posttrauma systemic inflammation. In this study, the effectiveness of cationic hyperbranched polyaminoglycosides derived from tobramycin (HPT) and disulfide-included HPT (ss-HPT) in scavenging cfNAs to mitigate posttrauma inflammation and hypercoagulation is investigated. Both cationic polymers demonstrate the ability to suppress DAMP-induced toll-like receptor (TLR) activation, inflammatory cytokine secretion, and hypercoagulation by efficiently scavenging cfNAs. Additionally, HPT and ss-HPT exhibit potent antibacterial efficacy attributed to the presence of tobramycin in their chemical composition. Furthermore, HPT and ss-HPT exhibit favorable modulatory effects on inflammation and therapeutic outcomes in a cecal ligation puncture (CLP) mouse abdominal trauma model. Notably, in vivo studies reveal that ss-HPT displayed high accumulation and retention in injured organs of traumatized mice while maintaining a higher biodegradation rate in healthy mice, contrasting with findings for HPT. Thus, functionalized ss-HPT, a bioreducible polyaminoglycoside, holds promise as an effective option to enhance therapeutic outcomes for trauma patients by alleviating posttrauma inflammation and coagulation complications.
Collapse
Affiliation(s)
- Yongqiang Xiao
- Department of Burn Surgerythe First Affiliated HospitalNaval Medical UniversityShanghai200433P. R. China
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- ENT InstituteDepartment of Facial Plastic and Reconstructive SurgeryEye & ENT HospitalFudan UniversityShanghai200031P. R. China
| | - He Fang
- Department of Burn Surgerythe First Affiliated HospitalNaval Medical UniversityShanghai200433P. R. China
| | - Yuefei Zhu
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Jie Zhou
- Department of Breast SurgeryAffiliated Cancer Hospital and InstituteGuangzhou Medical UniversityGuangzhou510095P. R. China
| | - Zhanzhan Dai
- Department of Burn Surgerythe First Affiliated HospitalNaval Medical UniversityShanghai200433P. R. China
| | - Hongxia Wang
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Zhaofan Xia
- Department of Burn Surgerythe First Affiliated HospitalNaval Medical UniversityShanghai200433P. R. China
| | - Zhaoxu Tu
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655P. R. China
| | - Kam W. Leong
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- Department of Systems BiologyColumbia University Medical CenterNew YorkNY10032USA
| |
Collapse
|
10
|
Bosco M, Romero R, Gallo DM, Suksai M, Gotsch F, Jung E, Chaemsaithong P, Tarca AL, Gomez-Lopez N, Arenas-Hernandez M, Meyyazhagan A, Al Qasem M, Franchi MP, Grossman LI, Aras S, Chaiworapongsa T. Clinical chorioamnionitis at term is characterized by changes in the plasma concentration of CHCHD2/MNRR1, a mitochondrial protein. J Matern Fetal Neonatal Med 2023; 36:2222333. [PMID: 37349086 PMCID: PMC10445405 DOI: 10.1080/14767058.2023.2222333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/24/2023]
Abstract
OBJECTIVE Mitochondrial dysfunction was observed in acute systemic inflammatory conditions such as sepsis and might be involved in sepsis-induced multi-organ failure. Coiled-Coil-Helix-Coiled-Coil-Helix Domain Containing 2 (CHCHD2), also known as Mitochondrial Nuclear Retrograde Regulator 1 (MNRR1), a bi-organellar protein located in the mitochondria and the nucleus, is implicated in cell respiration, survival, and response to tissue hypoxia. Recently, the reduction of the cellular CHCHD2/MNRR1 protein, as part of mitochondrial dysfunction, has been shown to play a role in the amplification of inflammatory cytokines in a murine model of lipopolysaccharide-induced systemic inflammation. The aim of this study was to determine whether the plasma concentration of CHCHD2/MNRR1 changed during human normal pregnancy, spontaneous labor at term, and clinical chorioamnionitis at term. METHODS We conducted a cross-sectional study that included the following groups: 1) non-pregnant women (n = 17); 2) normal pregnant women at various gestational ages from the first trimester until term (n = 110); 3) women at term with spontaneous labor (n = 50); and 4) women with clinical chorioamnionitis at term in labor (n = 25). Plasma concentrations of CHCHD2/MNRR1 were assessed by an enzyme-linked immunosorbent assay. RESULTS 1) Pregnant women at term in labor with clinical chorioamnionitis had a significantly higher plasma CHCHD2/MNRR1 concentration than those in labor without chorioamnionitis (p = .003); 2) CHCHD2/MNRR1 is present in the plasma of healthy non-pregnant and normal pregnant women without significant differences in its plasma concentrations between the two groups; 3) there was no correlation between maternal plasma CHCHD2/MNRR1 concentration and gestational age at venipuncture; and 4) plasma CHCHD2/MNRR1 concentration was not significantly different in women at term in spontaneous labor compared to those not in labor. CONCLUSIONS CHCHD2/MNRR1 is physiologically present in the plasma of healthy non-pregnant and normal pregnant women, and its concentration does not change with gestational age and parturition at term. However, plasma CHCHD2/MNRR1 is elevated in women at term with clinical chorioamnionitis. CHCHD2/MNRR1, a novel bi-organellar protein located in the mitochondria and the nucleus, is released into maternal plasma during systemic inflammation.
Collapse
Affiliation(s)
- Mariachiara Bosco
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, AOUI Verona, University of Verona, Verona, Italy
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Dahiana M Gallo
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Gynecology and Obstetrics, Universidad del Valle, Cali, Colombia
| | - Manaphat Suksai
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Francesca Gotsch
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Piya Chaemsaithong
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Mahidol University, Bangkok, Thailand
| | - Adi L Tarca
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Arun Meyyazhagan
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Centre of Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Malek Al Qasem
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Mutah University, Al-Karak, Jordan
| | - Massimo P Franchi
- Department of Obstetrics and Gynecology, AOUI Verona, University of Verona, Verona, Italy
| | - Lawrence I Grossman
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Siddhesh Aras
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
11
|
Janko J, Bečka E, Kmeťová K, Hudecová L, Konečná B, Celec P, Bajaj-Elliott M, Pastorek M. Neutrophil extracellular traps formation and clearance is enhanced in fever and attenuated in hypothermia. Front Immunol 2023; 14:1257422. [PMID: 37849757 PMCID: PMC10577177 DOI: 10.3389/fimmu.2023.1257422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/15/2023] [Indexed: 10/19/2023] Open
Abstract
Fever and hypothermia represent two opposite strategies for fighting systemic inflammation. Fever results in immune activation; hypothermia is associated with energy conservation. Systemic Inflammatory Response Syndrome (SIRS) remains a significant cause of mortality worldwide. SIRS can lead to a broad spectrum of clinical symptoms but importantly, patients can develop fever or hypothermia. During infection, polymorphonuclear cells (PMNs) such as neutrophils prevent pathogen dissemination through the formation of neutrophil extracellular traps (NETs) that ensnare and kill bacteria. However, when dysregulated, NETs also promote host tissue damage. Herein, we tested the hypothesis that temperature modulates NETs homeostasis in response to infection and inflammation. NETs formation was studied in response to infectious (Escherichia coli, Staphylococcus aureus) and sterile (mitochondria) agents. When compared to body temperature (37°C), NETs formation increased at 40°C; interestingly, the response was stunted at 35°C and 42°C. While CD16+ CD49d+ PMNs represent a small proportion of the neutrophil population, they formed ~45-85% of NETs irrespective of temperature. Temperature increased formyl peptide receptor 1 (FPR1) expression to a differential extent in CD16+ CD49d- vs. CD49d+ PMNSs, suggesting further complexity to neutrophil function in hypo/hyperthermic conditions. The capacity of NETs to induce Toll-like receptor 9 (TLR9)-mediated NF-κB activation was found to be temperature independent. Interestingly, NET degradation was enhanced at higher temperatures, which corresponded with greater plasma DNase activity in response to temperature increase. Collectively, our observations indicate that NETs formation and clearance are enhanced at 40°C whilst temperatures of 35°C and 42°C attenuate this response. Targeting PMN-driven immunity may represent new venues for intervention in pathological inflammation.
Collapse
Affiliation(s)
- Jakub Janko
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Emil Bečka
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Katarína Kmeťová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Letícia Hudecová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Barbora Konečná
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Peter Celec
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Mona Bajaj-Elliott
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Michal Pastorek
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
12
|
Picca A, Guerra F, Calvani R, Coelho-Júnior HJ, Landi F, Bucci C, Marzetti E. Mitochondrial-Derived Vesicles: The Good, the Bad, and the Ugly. Int J Mol Sci 2023; 24:13835. [PMID: 37762138 PMCID: PMC10531235 DOI: 10.3390/ijms241813835] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Mitophagy is crucial for maintaining mitochondrial quality. However, its assessment in vivo is challenging. The endosomal-lysosomal system is a more accessible pathway through which subtypes of extracellular vesicles (EVs), which also contain mitochondrial constituents, are released for disposal. The inclusion of mitochondrial components into EVs occurs in the setting of mild mitochondrial damage and during impairment of lysosomal function. By releasing mitochondrial-derived vesicles (MDVs), cells limit the unload of mitochondrial damage-associated molecular patterns with proinflammatory activity. Both positive and negative effects of EVs on recipient cells have been described. Whether this is due to the production of EVs other than those containing mitochondria, such as MDVs, holding specific biological functions is currently unknown. Evidence on the existence of different MDV subtypes has been produced. However, their characterization is not always pursued, which would be relevant to exploring the dynamics of mitochondrial quality control in health and disease. Furthermore, MDV classification may be instrumental in understanding their biological roles and promoting their implementation as biomarkers in clinical studies.
Collapse
Affiliation(s)
- Anna Picca
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy;
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (F.L.); (E.M.)
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, 73100 Lecce, Italy; (F.G.); (C.B.)
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (F.L.); (E.M.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Hélio José Coelho-Júnior
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (F.L.); (E.M.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, 73100 Lecce, Italy; (F.G.); (C.B.)
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (F.L.); (E.M.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| |
Collapse
|
13
|
Bello S, Lasierra AB, López-Vergara L, de Diego C, Torralba L, de Gopegui PR, Lahoz R, Abadía C, Godino J, Cebollada A, Jimeno B, Bello C, Tejada A, Torres A. IL-6 and cfDNA monitoring throughout COVID-19 hospitalization are accurate markers of its outcomes. Respir Res 2023; 24:125. [PMID: 37147677 PMCID: PMC10161166 DOI: 10.1186/s12931-023-02426-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 04/18/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Severe COVID-19 entails a dysregulated immune response, most likely inflammation related to a lack of virus control. A better understanding of immune toxicity, immunosuppression balance, and COVID-19 assessments could help determine whether different clinical presentations are driven by specific types of immune responses. The progression of the immune response and tissular damage could predict outcomes and may help in the management of patients. METHODS We collected 201 serum samples from 93 hospitalised patients classified as moderately, severely, and critically ill. We differentiated the viral, early inflammatory, and late inflammatory phases and included 72 patients with 180 samples in separate stages for longitudinal study and 55 controls. We studied selected cytokines, P-selectin, and the tissue damage markers lactate dehydrogenase (LDH) and cell-free DNA (cfDNA). RESULTS TNF-α, IL-6, IL-8, and G-CSF were associated with severity and mortality, but only IL-6 increased since admission in the critical patients and non-survivors, correlating with damage markers. The lack of a significant decrease in IL-6 levels in the critical patients and non-survivors in the early inflammatory phase (a decreased presence in the other patients) suggests that these patients did not achieve viral control on days 10-16. For all patients, lactate dehydrogenase and cfDNA levels increased with severity, and cfDNA levels increased in the non-survivors from the first sample (p = 0.002) to the late inflammatory phase (p = 0.031). In the multivariate study, cfDNA was an independent risk factor for mortality and ICU admission. CONCLUSIONS The distinct progression of IL-6 levels in the course of the disease, especially on days 10-16, was a good marker of progression to critical status and mortality and could guide the start of IL-6 blockade. cfDNA was an accurate marker of severity and mortality from admission and throughout COVID-19 progression.
Collapse
Affiliation(s)
- Salvador Bello
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel La Católica 1-9, 50009, Zaragoza, Spain.
| | | | - Lucía López-Vergara
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel La Católica 1-9, 50009, Zaragoza, Spain
| | - Cristina de Diego
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel La Católica 1-9, 50009, Zaragoza, Spain
| | - Laura Torralba
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel La Católica 1-9, 50009, Zaragoza, Spain
| | | | - Raquel Lahoz
- Department of Biochemistry, Miguel Servet University Hospital, Zaragoza, Spain
| | - Claudia Abadía
- Department of Biochemistry, Miguel Servet University Hospital, Zaragoza, Spain
| | - Javier Godino
- Department of Cytometry and Cell Separation, Aragon Institute of Health Sciences (IACS), Zaragoza, Spain
| | - Alberto Cebollada
- Biocomputing Technical Scientific Service, Aragon Institute of Health Sciences (IACS), Zaragoza, Spain
| | - Beatriz Jimeno
- Department of Cytometry and Cell Separation, Aragon Institute of Health Sciences (IACS), Zaragoza, Spain
| | - Carlota Bello
- Department of Radiology, Hospital Clínico Lozano Blesa, Zaragoza, Spain
| | - Antonio Tejada
- Intensive Care Unit, Miguel Servet University Hospital, Zaragoza, Spain
| | - Antoni Torres
- Servei de Pneumologia, Hospital Clinic, Universitat de Barcelona, IDIBAPS, ICREA, CIBERESUCICOVID, Barcelona, Spain
| |
Collapse
|
14
|
Chaiyarit S, Thongboonkerd V. Mitochondria-derived vesicles and their potential roles in kidney stone disease. J Transl Med 2023; 21:294. [PMID: 37131163 PMCID: PMC10152607 DOI: 10.1186/s12967-023-04133-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/14/2023] [Indexed: 05/04/2023] Open
Abstract
Recent evidence has shown significant roles of mitochondria-derived vesicles (MDVs) in mitochondrial quality control (MQC) system. Under mild stress condition, MDVs are formed to carry the malfunctioned mitochondrial components, such as mitochondrial DNA (mtDNA), peptides, proteins and lipids, to be eliminated to restore normal mitochondrial structure and functions. Under severe oxidative stress condition, mitochondrial dynamics (fission/fusion) and mitophagy are predominantly activated to rescue mitochondrial structure and functions. Additionally, MDVs generation can be also triggered as the major MQC machinery to cope with unhealthy mitochondria when mitophagy is unsuccessful for eliminating the damaged mitochondria or mitochondrial fission/fusion fail to recover the mitochondrial structure and functions. This review summarizes the current knowledge on MDVs and discuss their roles in physiologic and pathophysiologic conditions. In addition, the potential clinical relevance of MDVs in therapeutics and diagnostics of kidney stone disease (KSD) are emphasized.
Collapse
Affiliation(s)
- Sakdithep Chaiyarit
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 6th Floor, SiMR Building, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 6th Floor, SiMR Building, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand.
| |
Collapse
|
15
|
Fricchione G. Mind body medicine: a modern bio-psycho-social model forty-five years after Engel. Biopsychosoc Med 2023; 17:12. [PMID: 36997979 PMCID: PMC10060142 DOI: 10.1186/s13030-023-00268-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 03/31/2023] Open
Affiliation(s)
- Gregory Fricchione
- Benson-Henry Institute for Mind Body Medicine at Massachusetts General Hospital, Boston, USA.
- Department of Psychiatry, Massachusetts General Hospital, Boston, USA.
- Harvard Medical School, Boston, USA.
| |
Collapse
|
16
|
Skulachev VP, Vyssokikh MY, Chernyak BV, Averina OA, Andreev-Andrievskiy AA, Zinovkin RA, Lyamzaev KG, Marey MV, Egorov MV, Frolova OJ, Zorov DB, Skulachev MV, Sadovnichii VA. Mitochondrion-targeted antioxidant SkQ1 prevents rapid animal death caused by highly diverse shocks. Sci Rep 2023; 13:4326. [PMID: 36922552 PMCID: PMC10017827 DOI: 10.1038/s41598-023-31281-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
The response to stress involves the activation of pathways leading either to protection from the stress origin, eventually resulting in development of stress resistance, or activation of the rapid death of the organism. Here we hypothesize that mitochondrial reactive oxygen species (mtROS) play a key role in stress-induced programmed death of the organism, which we called "phenoptosis" in 1997. We demonstrate that the synthetic mitochondria-targeted antioxidant SkQ1 (which specifically abolishes mtROS) prevents rapid death of mice caused by four mechanistically very different shocks: (a) bacterial lipopolysaccharide (LPS) shock, (b) shock in response to intravenous mitochondrial injection, (c) cold shock, and (d) toxic shock caused by the penetrating cation C12TPP. Importantly, under all these stresses mortality was associated with a strong elevation of the levels of pro-inflammatory cytokines and administration of SkQ1 was able to switch off the cytokine storms. Since the main effect of SkQ1 is the neutralization of mtROS, this study provides evidence for the role of mtROS in the activation of innate immune responses mediating stress-induced death of the organism. We propose that SkQ1 may be used clinically to support patients in critical conditions, such as septic shock, extensive trauma, cooling, and severe infection by bacteria or viruses.
Collapse
Affiliation(s)
- V P Skulachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.
| | - M Yu Vyssokikh
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.
| | - B V Chernyak
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991. .,Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991.
| | - O A Averina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.,Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - A A Andreev-Andrievskiy
- Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991.,Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - R A Zinovkin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.,Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - K G Lyamzaev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.,Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - M V Marey
- Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia, 117198
| | - M V Egorov
- Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - O J Frolova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.,Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - D B Zorov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - M V Skulachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - V A Sadovnichii
- Faculty of Mechanics and Mathematics, Lomonosov Moscow State University, Moscow, Russia, 119991
| |
Collapse
|
17
|
Tiwari-Heckler S, Lee GR, Harbison J, Ledderose C, Csizmadia E, Melton D, Zhang Q, Junger W, Chen G, Hauser CJ, Otterbein LE, Longhi MS, Robson SC. Extracellular mitochondria drive CD8 T cell dysfunction in trauma by upregulating CD39. Thorax 2023; 78:151-159. [PMID: 35613855 PMCID: PMC9691787 DOI: 10.1136/thoraxjnl-2021-218047] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 04/04/2022] [Indexed: 01/21/2023]
Abstract
RATIONALE The increased mortality and morbidity seen in critically injured patients appears associated with systemic inflammatory response syndrome (SIRS) and immune dysfunction, which ultimately predisposes to infection. Mitochondria released by injury could generate danger molecules, for example, ATP, which in turn would be rapidly scavenged by ectonucleotidases, expressed on regulatory immune cells. OBJECTIVE To determine the association between circulating mitochondria, purinergic signalling and immune dysfunction after trauma. METHODS We tested the impact of hepatocyte-derived free mitochondria on blood-derived and lung-derived CD8 T cells in vitro and in experimental mouse models in vivo. In parallel, immune phenotypic analyses were conducted on blood-derived CD8 T cells obtained from trauma patients. RESULTS Isolated intact mitochondria are functional and generate ATP ex vivo. Extracellular mitochondria perturb CD8+ T cells in co-culture, inducing select features of immune exhaustion in vitro. These effects are modulated by scavenging ATP, modelled by addition of apyrase in vitro. Injection of intact mitochondria into recipient mice markedly upregulates the ectonucleotidase CD39, and other immune checkpoint markers in circulating CD8+ T cells. We note that mice injected with mitochondria, prior to instilling bacteria into the lung, exhibit more severe lung injury, characterised by elevated neutrophil influx and by changes in CD8+ T cell cytotoxic capacity. Importantly, the development of SIRS in injured humans, is likewise associated with disordered purinergic signalling and CD8 T cell dysfunction. CONCLUSION These studies in experimental models and in a cohort of trauma patients reveal important associations between extracellular mitochondria, aberrant purinergic signalling and immune dysfunction. These pathogenic factors with immune exhaustion are linked to SIRS and could be targeted therapeutically.
Collapse
Affiliation(s)
- Shilpa Tiwari-Heckler
- Gastroenterology, University Hospital Heidelberg Medical Clinic, Heidelberg, Germany
- Center for Inflammation Research, Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ghee Rye Lee
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - James Harbison
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Eva Csizmadia
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - David Melton
- Center for Inflammation Research, Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Wolfgang Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Guanqing Chen
- Center for Inflammation Research, Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Carl J Hauser
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Leo E Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Serena Longhi
- Center for Inflammation Research, Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Simon Christopher Robson
- Center for Inflammation Research, Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Tu H, Li YL. Inflammation balance in skeletal muscle damage and repair. Front Immunol 2023; 14:1133355. [PMID: 36776867 PMCID: PMC9909416 DOI: 10.3389/fimmu.2023.1133355] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
Responding to tissue injury, skeletal muscles undergo the tissue destruction and reconstruction accompanied with inflammation. The immune system recognizes the molecules released from or exposed on the damaged tissue. In the local minor tissue damage, tissue-resident macrophages sequester pro-inflammatory debris to prevent initiation of inflammation. In most cases of the skeletal muscle injury, however, a cascade of inflammation will be initiated through activation of local macrophages and mast cells and recruitment of immune cells from blood circulation to the injured site by recongnization of damage-associated molecular patterns (DAMPs) and activated complement system. During the inflammation, macrophages and neutrophils scavenge the tissue debris to release inflammatory cytokines and the latter stimulates myoblast fusion and vascularization to promote injured muscle repair. On the other hand, an abundance of released inflammatory cytokines and chemokines causes the profound hyper-inflammation and mobilization of immune cells to trigger a vicious cycle and lead to the cytokine storm. The cytokine storm results in the elevation of cytolytic and cytotoxic molecules and reactive oxygen species (ROS) in the damaged muscle to aggravates the tissue injury, including the healthy bystander tissue. Severe inflammation in the skeletal muscle can lead to rhabdomyolysis and cause sepsis-like systemic inflammation response syndrome (SIRS) and remote organ damage. Therefore, understanding more details on the involvement of inflammatory factors and immune cells in the skeletal muscle damage and repair can provide the new precise therapeutic strategies, including attenuation of the muscle damage and promotion of the muscle repair.
Collapse
|
19
|
Chen L, Zhang X, Liu Y, Liu L, Liang X, Yang S, Xia Q, Jin T, Ma Y, Chen Y, Yuan X, Tie Y, Gu Y, Fang C, Chen S, Mo F, Yu T, Hu Y, Qian Z, Peng Y, Geng J, Zhou Z, Wu M, Ding J, Yang D, Wei X. JMJD3 Is Required for Acute Pancreatitis and Pancreatitis-Associated Lung Injury. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:180-190. [PMID: 36458991 PMCID: PMC9772398 DOI: 10.4049/jimmunol.2200484] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/01/2022] [Indexed: 01/04/2023]
Abstract
Acute pancreatitis (AP) can be complicated by inflammatory disorders of remote organs, such as lung injury, in which Jumonji domain-containing protein 3 (JMJD3) plays a vital role in proinflammatory responses. Currently, we found that JMJD3 expression was upregulated in the pancreas and lung in an AP male mouse model, which was also confirmed in AP patients. Further experiments revealed that the upregulation of JMJD3 and proinflammatory effects were possibly exerted by mitochondrial DNA (mtDNA) or oxidized-mtDNA from tissue injury caused by AP. The release of mtDNA and oxidized-mtDNA contributed to the infiltration of inflammatory monocytes in lung injury through the stimulator of IFN genes (STING)/TLR9-NF-κB-JMJD3-TNF-α pathway. The inhibition of JMJD3 or utilization of Jmjd3-cKO mice significantly alleviated pulmonary inflammation induced by AP. Blocking mtDNA oxidation or knocking down the TLR9/STING pathway effectively alleviated inflammation. Therefore, inhibition of JMJD3 or STING/TLR9 pathway blockage might be a potential therapeutic strategy to treat AP and the associated lung injury.
Collapse
Affiliation(s)
- Li Chen
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Xiangxian Zhang
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yu Liu
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Li Liu
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Xiao Liang
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Shengqun Yang
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Qing Xia
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Tao Jin
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yun Ma
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yonghua Chen
- Department of Pancreatic Surgery/Pancreatic Disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Xia Yuan
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yan Tie
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yangzhuo Gu
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Chunju Fang
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Siyuan Chen
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Fei Mo
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Ting Yu
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yuzhu Hu
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zhiyong Qian
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yong Peng
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Jia Geng
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zongguang Zhou
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Min Wu
- Department of Biochemistry and Molecular Biology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND; and
| | - Jiansheng Ding
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Daoke Yang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiawei Wei
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
20
|
Tiwari-Heckler S, Robson SC, Longhi MS. Mitochondria Drive Immune Responses in Critical Disease. Cells 2022; 11:cells11244113. [PMID: 36552877 PMCID: PMC9777392 DOI: 10.3390/cells11244113] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Mitochondria engage in multiple cellular and extracellular signaling pathways ranging from metabolic control, antiviral and antibacterial host defense to the modulation of inflammatory responses following cellular damage and stress. The remarkable contributions of these organelles to innate and adaptive immunity, shape cell phenotype and modulate their functions during infection, after trauma and in the setting of inflammatory disease. We review the latest knowledge of mitochondrial biology and then discuss how these organelles may impact immune cells to drive aberrant immune responses in critical disease.
Collapse
Affiliation(s)
- Shilpa Tiwari-Heckler
- Department of Gastroenterology, University Hospital Heidelberg Medical Clinic, 69120 Heidelberg, Germany
| | - Simon C. Robson
- Center for Inflammation Research, Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Maria Serena Longhi
- Center for Inflammation Research, Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Correspondence:
| |
Collapse
|
21
|
Nunns GR, Vigneshwar N, Kelher MR, Stettler GR, Gera L, Reisz JA, D’Alessandro A, Ryon J, Hansen KC, Burke T, Gamboni F, Moore EE, Peltz ED, Cohen MJ, Jones KL, Sauaia A, Liang X, Banerjee A, Ghasabyan A, Chandler JG, Rodawig S, Jones C, Eitel A, Hom P, Silliman CC. Succinate Activation of SUCNR1 Predisposes Severely Injured Patients to Neutrophil-mediated ARDS. Ann Surg 2022; 276:e944-e954. [PMID: 33214479 PMCID: PMC8128932 DOI: 10.1097/sla.0000000000004644] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Identify the metabolites that are increased in the plasma of severely injured patients that developed ARDS versus severely injured patients that did not, and assay if these increased metabolites prime pulmonary sequestration of neutrophils (PMNs) and induce pulmonary sequestration in an animal model of ARDS. We hypothesize that metabolic derangement due to advanced shock in critically injured patients leads to the PMNs, which serves as the first event in the ARDS. Summary of Background Data: Intracellular metabolites accumulate in the plasma of severely injured patients. METHODS Untargeted metabolomics profiling of 67 critically injured patients was completed to establish a metabolic signature associated with ARDS development. Metabolites that significantly increased were assayed for PMN priming activity in vitro. The metabolites that primed PMNs were tested in a 2-event animal model of ARDS to identify a molecular link between circulating metabolites and clinical risk for ARDS. RESULTS After controlling for confounders, 4 metabolites significantly increased: creatine, dehydroascorbate, fumarate, and succinate in trauma patients who developed ARDS ( P < 0.05). Succinate alone primed the PMN oxidase in vitro at physiologically relevant levels. Intravenous succinate-induced PMN sequestration in the lung, a first event, and followed by intravenous lipopolysaccharide, a second event, resulted in ARDS in vivo requiring PMNs. SUCNR1 inhibition abrogated PMN priming, PMN sequestration, and ARDS. Conclusion: Significant increases in plasma succinate post-injury may serve as the first event in ARDS. Targeted inhibition of the SUCNR1 may decrease ARDS development from other disease states to prevent ARDS globally.
Collapse
Affiliation(s)
- Geoffrey R Nunns
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Navin Vigneshwar
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Marguerite R Kelher
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
- Vitalant Research Institute, Vitalant Denver, Denver, CO
| | - Gregory R Stettler
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Lajos Gera
- Biochemistry and Molecular Genetics, School of Medicine University of Colorado, Aurora, CO
| | - Julie A. Reisz
- Biochemistry and Molecular Genetics, School of Medicine University of Colorado, Aurora, CO
| | - Angelo D’Alessandro
- Biochemistry and Molecular Genetics, School of Medicine University of Colorado, Aurora, CO
| | - Joshua Ryon
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Kirk C Hansen
- Biochemistry and Molecular Genetics, School of Medicine University of Colorado, Aurora, CO
| | - Timothy Burke
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
- Vitalant Research Institute, Vitalant Denver, Denver, CO
| | - Fabia Gamboni
- Biochemistry and Molecular Genetics, School of Medicine University of Colorado, Aurora, CO
| | - Ernest E. Moore
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
- Department of Surgery, Denver Health Medical Center, Denver, CO
| | - Erik D Peltz
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Mitchell J Cohen
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
- Department of Surgery, Denver Health Medical Center, Denver, CO
| | | | - Angela Sauaia
- Department of Surgery, Denver Health Medical Center, Denver, CO
- School of Public Health, University of Colorado, Aurora, CO
| | - Xiayuan Liang
- Pathology, School of Medicine, University of Colorado, Aurora, CO
| | - Anirban Banerjee
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Arsen Ghasabyan
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - James G Chandler
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Sophia Rodawig
- Vitalant Research Institute, Vitalant Denver, Denver, CO
- College of Arts and Letters, University of Notre Dame, Notre Dame, IL
| | - Carter Jones
- Vitalant Research Institute, Vitalant Denver, Denver, CO
- College of Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Andrew Eitel
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Patrick Hom
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Christopher C Silliman
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
- Pediatrics, School of Medicine University of Colorado, CO
- Vitalant Research Institute, Vitalant Denver, Denver, CO
| |
Collapse
|
22
|
Műzes G, Bohusné Barta B, Szabó O, Horgas V, Sipos F. Cell-Free DNA in the Pathogenesis and Therapy of Non-Infectious Inflammations and Tumors. Biomedicines 2022; 10:2853. [PMID: 36359370 PMCID: PMC9687442 DOI: 10.3390/biomedicines10112853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022] Open
Abstract
The basic function of the immune system is the protection of the host against infections, along with the preservation of the individual antigenic identity. The process of self-tolerance covers the discrimination between self and foreign antigens, including proteins, nucleic acids, and larger molecules. Consequently, a broken immunological self-tolerance results in the development of autoimmune or autoinflammatory disorders. Immunocompetent cells express pattern-recognition receptors on their cell membrane and cytoplasm. The majority of endogenous DNA is located intracellularly within nuclei and mitochondria. However, extracellular, cell-free DNA (cfDNA) can also be detected in a variety of diseases, such as autoimmune disorders and malignancies, which has sparked interest in using cfDNA as a possible biomarker. In recent years, the widespread use of liquid biopsies and the increasing demand for screening, as well as monitoring disease activity and therapy response, have enabled the revival of cfDNA research. The majority of studies have mainly focused on the function of cfDNA as a biomarker. However, research regarding the immunological consequences of cfDNA, such as its potential immunomodulatory or therapeutic benefits, is still in its infancy. This article discusses the involvement of various DNA-sensing receptors (e.g., absent in melanoma-2; Toll-like receptor 9; cyclic GMP-AMP synthase/activator of interferon genes) in identifying host cfDNA as a potent danger-associated molecular pattern. Furthermore, we aim to summarize the results of the experimental studies that we recently performed and highlight the immunomodulatory capacity of cfDNA, and thus, the potential for possible therapeutic consideration.
Collapse
Affiliation(s)
| | | | | | | | - Ferenc Sipos
- Department of Internal Medicine and Hematology, Semmelweis University, Szentkirályi Street 46, 1088 Budapest, Hungary
| |
Collapse
|
23
|
Dobson GP, Morris JL, Letson HL. Why are bleeding trauma patients still dying? Towards a systems hypothesis of trauma. Front Physiol 2022; 13:990903. [PMID: 36148305 PMCID: PMC9485567 DOI: 10.3389/fphys.2022.990903] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/12/2022] [Indexed: 12/14/2022] Open
Abstract
Over the years, many explanations have been put forward to explain early and late deaths following hemorrhagic trauma. Most include single-event, sequential contributions from sympathetic hyperactivity, endotheliopathy, trauma-induced coagulopathy (TIC), hyperinflammation, immune dysfunction, ATP deficit and multiple organ failure (MOF). We view early and late deaths as a systems failure, not as a series of manifestations that occur over time. The traditional approach appears to be a by-product of last century's highly reductionist, single-nodal thinking, which also extends to patient management, drug treatment and drug design. Current practices appear to focus more on alleviating symptoms rather than addressing the underlying problem. In this review, we discuss the importance of the system, and focus on the brain's "privilege" status to control secondary injury processes. Loss of status from blood brain barrier damage may be responsible for poor outcomes. We present a unified Systems Hypothesis Of Trauma (SHOT) which involves: 1) CNS-cardiovascular coupling, 2) Endothelial-glycocalyx health, and 3) Mitochondrial integrity. If central control of cardiovascular coupling is maintained, we hypothesize that the endothelium will be protected, mitochondrial energetics will be maintained, and immune dysregulation, inflammation, TIC and MOF will be minimized. Another overlooked contributor to early and late deaths following hemorrhagic trauma is from the trauma of emergent surgery itself. This adds further stress to central control of secondary injury processes. New point-of-care drug therapies are required to switch the body's genomic and proteomic programs from an injury phenotype to a survival phenotype. Currently, no drug therapy exists that targets the whole system following major trauma.
Collapse
Affiliation(s)
- Geoffrey P. Dobson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia
| | | | | |
Collapse
|
24
|
Dobson GP, Morris JL, Letson HL. Immune dysfunction following severe trauma: A systems failure from the central nervous system to mitochondria. Front Med (Lausanne) 2022; 9:968453. [PMID: 36111108 PMCID: PMC9468749 DOI: 10.3389/fmed.2022.968453] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/01/2022] [Indexed: 12/20/2022] Open
Abstract
When a traumatic injury exceeds the body's internal tolerances, the innate immune and inflammatory systems are rapidly activated, and if not contained early, increase morbidity and mortality. Early deaths after hospital admission are mostly from central nervous system (CNS) trauma, hemorrhage and circulatory collapse (30%), and later deaths from hyperinflammation, immunosuppression, infection, sepsis, acute respiratory distress, and multiple organ failure (20%). The molecular drivers of secondary injury include damage associated molecular patterns (DAMPs), pathogen associated molecular patterns (PAMPs) and other immune-modifying agents that activate the hypothalamic-pituitary-adrenal (HPA) axis and sympathetic stress response. Despite a number of drugs targeting specific anti-inflammatory and immune pathways showing promise in animal models, the majority have failed to translate. Reasons for failure include difficulty to replicate the heterogeneity of humans, poorly designed trials, inappropriate use of specific pathogen-free (SPF) animals, ignoring sex-specific differences, and the flawed practice of single-nodal targeting. Systems interconnectedness is a major overlooked factor. We argue that if the CNS is protected early after major trauma and control of cardiovascular function is maintained, the endothelial-glycocalyx will be protected, sufficient oxygen will be delivered, mitochondrial energetics will be maintained, inflammation will be resolved and immune dysfunction will be minimized. The current challenge is to develop new systems-based drugs that target the CNS coupling of whole-body function.
Collapse
Affiliation(s)
- Geoffrey P. Dobson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia
| | | | | |
Collapse
|
25
|
Mahmoodpoor A, Sanaie S, Ostadi Z, Eskandari M, Behrouzi N, Asghari R, Zahirnia A, Sohrabifar N, Kazeminasab S. Roles of mitochondrial DNA in dynamics of the immune response to COVID-19. Gene 2022; 836:146681. [PMID: 35728769 PMCID: PMC9219426 DOI: 10.1016/j.gene.2022.146681] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/29/2022] [Accepted: 06/10/2022] [Indexed: 12/18/2022]
Abstract
Mitochondria dynamics have a pivotal role in many aspects of immune function. Viral infections affect mitochondrial dynamics and trigger the release of mitochondrial DNA (mtDNA) in host cells. Released mtDNA guides the immune response towards an inflammatory response against pathogens. In addition, circulating cell-free mtDNA (ccf-mtDNA) is considered an invaluable indicator for the prognosis and severity of infectious diseases. This study provides an overview of the role of mtDNA in the dynamics of the immune response to COVID-19. We focused on the possible roles of mtDNA in inducing the signaling pathways, and the inflammasome activation and regulation in SARS-CoV-2. Targeting mtDNA-related pathways can provide critical insights into therapeutic strategies for COVID-19.
Collapse
Affiliation(s)
- Ata Mahmoodpoor
- Research Center for Integrative Medicine in Aging, Aging research institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sarvin Sanaie
- Research Center for Integrative Medicine in Aging, Aging research institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zoherh Ostadi
- Department of Anesthesiology and intensive care, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maqsoud Eskandari
- Department of Anesthesiology and intensive care, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Behrouzi
- Department of Anesthesiology and intensive care, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roqayyeh Asghari
- Department of Anesthesiology and intensive care, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Zahirnia
- Nasle Farda Health Foundation, Medical Genetic Laboratory, Tabriz, Iran
| | - Nasim Sohrabifar
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Somayeh Kazeminasab
- Nasle Farda Health Foundation, Medical Genetic Laboratory, Tabriz, Iran; Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
26
|
Zambrano K, Barba D, Castillo K, Robayo P, Argueta-Zamora D, Sanon S, Arizaga E, Caicedo A, Gavilanes AWD. The war against Alzheimer, the mitochondrion strikes back! Mitochondrion 2022; 64:125-135. [PMID: 35337984 DOI: 10.1016/j.mito.2022.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/08/2022] [Accepted: 03/21/2022] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a leading neurodegenerative pathology associated with aging worldwide. It is estimated that AD prevalence will increase from 5.8 million people today to 13.8 million by 2050 in the United States alone. AD effects in the brain are well known; however, there is still a lack of knowledge about the cellular mechanisms behind the origin of AD. It is known that AD induces cellular stress affecting the energy metabolism in brain cells. During the pathophysiological advancement of AD, damaged mitochondria enter a vicious cycle, producing reactive oxygen species (ROS), harming mitochondrial DNA and proteins, leading to more ROS and cellular death. Additionally, mitochondria are interconnected with the plaques formed by amyloid-β in AD and have underlying roles in the progression of the disease and severity. For years, the biomedical field struggled to develop new therapeutic options for AD without a significant advancement. However, mitochondria are striking back existing outside cells in a new mechanism of intercellular communication. Extracellular mitochondria are exchanged from healthy to damaged cells to rescue those with a perturbed metabolism in a process that could be applied as a new therapeutic option to repair those brain cells affected by AD. In this review we highlight key aspects of mitochondria's role in CNS' physiology and neurodegenerative disorders, focusing on AD. We also suggest how mitochondria strikes back as a therapeutic target and as a potential agent to be transplanted to repair neurons affected by AD.
Collapse
Affiliation(s)
- Kevin Zambrano
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, 17-12-841, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina, 17-12-841, Quito, Ecuador; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands; Mito-Act Research Consortium, Quito, Ecuador; Instituto de Neurociencias, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Diego Barba
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, 17-12-841, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina, 17-12-841, Quito, Ecuador; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Karina Castillo
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, 17-12-841, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina, 17-12-841, Quito, Ecuador
| | - Paola Robayo
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, 17-12-841, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina, 17-12-841, Quito, Ecuador
| | | | | | - Eduardo Arizaga
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, 17-12-841, Quito, Ecuador
| | - Andres Caicedo
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, 17-12-841, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina, 17-12-841, Quito, Ecuador; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands; Mito-Act Research Consortium, Quito, Ecuador; Sistemas Médicos SIME, Universidad San Francisco de Quito, Quito, Ecuador
| | - Antonio W D Gavilanes
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, 17-12-841, Quito, Ecuador; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
27
|
Laudanski K. Quo Vadis Anesthesiologist? The Value Proposition of Future Anesthesiologists Lies in Preserving or Restoring Presurgical Health after Surgical Insult. J Clin Med 2022; 11:1135. [PMID: 35207406 PMCID: PMC8879076 DOI: 10.3390/jcm11041135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/18/2022] [Indexed: 12/26/2022] Open
Abstract
This Special Issue of the Journal of Clinical Medicine is devoted to anesthesia and perioperative care [...].
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA 19104, USA; ; Tel.: +1-215-662-8000
- Leonard Davis Institute for Healthcare Economics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Lee GR, Gallo D, Alves de Souza RW, Tiwari-Heckler S, Csizmadia E, Harbison JD, Shankar S, Banner-Goodspeed V, Yaffe MB, Longhi MS, Hauser CJ, Otterbein LE. Trauma-induced heme release increases susceptibility to bacterial infection. JCI Insight 2021; 6:e150813. [PMID: 34520397 PMCID: PMC8564912 DOI: 10.1172/jci.insight.150813] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/09/2021] [Indexed: 12/01/2022] Open
Abstract
Infection is a common complication of major trauma that causes significantly increased morbidity and mortality. The mechanisms, however, linking tissue injury to increased susceptibility to infection remain poorly understood. To study this relationship, we present a potentially novel murine model in which a major liver crush injury is followed by bacterial inoculation into the lung. We find that such tissue trauma both impaired bacterial clearance and was associated with significant elevations in plasma heme levels. While neutrophil (PMN) recruitment to the lung in response to Staphylococcus aureus was unchanged after trauma, PMN cleared bacteria poorly. Moreover, PMN show > 50% less expression of TLR2, which is responsible, in part, for bacterial recognition. Administration of heme effectively substituted for trauma. Finally, day 1 trauma patients (n = 9) showed similar elevations in free heme compared with that seen after murine liver injury, and circulating PMN showed similar TLR2 reduction compared with volunteers (n = 6). These findings correlate to high infection rates.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Valerie Banner-Goodspeed
- Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael B Yaffe
- Department of Surgery and.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Maria Serena Longhi
- Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
29
|
Bobrov AG, Getnet D, Swierczewski B, Jacobs A, Medina-Rojas M, Tyner S, Watters C, Antonic V. Evaluation of Pseudomonas aeruginosa pathogenesis and therapeutics in military-relevant animal infection models. APMIS 2021; 130:436-457. [PMID: 34132418 DOI: 10.1111/apm.13119] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/21/2021] [Indexed: 01/02/2023]
Abstract
Modern combat-related injuries are often associated with acute polytrauma. As a consequence of severe combat-related injuries, a dysregulated immune response results in serious infectious complications. The gram-negative bacterium Pseudomonas aeruginosa is an opportunistic pathogen that often causes life-threatening bloodstream, lung, bone, urinary tract, and wound infections following combat-related injuries. The rise in the number of multidrug-resistant P. aeruginosa strains has elevated its importance to civilian clinicians and military medicine. Development of novel therapeutics and treatment options for P. aeruginosa infections is urgently needed. During the process of drug discovery and therapeutic testing, in vivo testing in animal models is a critical step in the bench-to-bedside approach, and required for Food and Drug Administration approval. Here, we review current and past literature with a focus on combat injury-relevant animal models often used to understand infection development, the interplay between P. aeruginosa and the host, and evaluation of novel treatments. Specifically, this review focuses on the following animal infection models: wound, burn, bone, lung, urinary tract, foreign body, and sepsis.
Collapse
Affiliation(s)
- Alexander G Bobrov
- Wound Infections Department, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Derese Getnet
- Wound Infections Department, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Brett Swierczewski
- Wound Infections Department, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Anna Jacobs
- Wound Infections Department, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Maria Medina-Rojas
- Wound Infections Department, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Stuart Tyner
- US Army Medical Research and Development Command Military Infectious Diseases Research Program, Frederick, Maryland, USA
| | - Chase Watters
- Naval Medical Research Unit-3, Ghana Detachment, Accra, Ghana
| | - Vlado Antonic
- Wound Infections Department, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| |
Collapse
|
30
|
Andargie TE, Tsuji N, Seifuddin F, Jang MK, Yuen PS, Kong H, Tunc I, Singh K, Charya A, Wilkins K, Nathan S, Cox A, Pirooznia M, Star RA, Agbor-Enoh S. Cell-free DNA maps COVID-19 tissue injury and risk of death and can cause tissue injury. JCI Insight 2021; 6:147610. [PMID: 33651717 PMCID: PMC8119224 DOI: 10.1172/jci.insight.147610] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/02/2021] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION The clinical course of coronavirus 2019 (COVID-19) is heterogeneous, ranging from mild to severe multiorgan failure and death. In this study, we analyzed cell-free DNA (cfDNA) as a biomarker of injury to define the sources of tissue injury that contribute to such different trajectories. METHODS We conducted a multicenter prospective cohort study to enroll patients with COVID-19 and collect plasma samples. Plasma cfDNA was subject to bisulfite sequencing. A library of tissue-specific DNA methylation signatures was used to analyze sequence reads to quantitate cfDNA from different tissue types. We then determined the correlation of tissue-specific cfDNA measures to COVID-19 outcomes. Similar analyses were performed for healthy controls and a comparator group of patients with respiratory syncytial virus and influenza. RESULTS We found markedly elevated levels and divergent tissue sources of cfDNA in COVID-19 patients compared with patients who had influenza and/or respiratory syncytial virus and with healthy controls. The major sources of cfDNA in COVID-19 were hematopoietic cells, vascular endothelium, hepatocytes, adipocytes, kidney, heart, and lung. cfDNA levels positively correlated with COVID-19 disease severity, C-reactive protein, and D-dimer. cfDNA profile at admission identified patients who subsequently required intensive care or died during hospitalization. Furthermore, the increased cfDNA in COVID-19 patients generated excessive mitochondrial ROS (mtROS) in renal tubular cells in a concentration-dependent manner. This mtROS production was inhibited by a TLR9-specific antagonist. CONCLUSION cfDNA maps tissue injury that predicts COVID-19 outcomes and may mechanistically propagate COVID-19–induced tissue injury. FUNDING Intramural Targeted Anti–COVID-19 grant, NIH.
Collapse
Affiliation(s)
- Temesgen E Andargie
- Genomic Research Alliance for Transplantation (GRAfT) and Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA.,Department of Biology, Howard University, Washington DC, USA
| | - Naoko Tsuji
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | | | - Moon Kyoo Jang
- Genomic Research Alliance for Transplantation (GRAfT) and Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Peter St Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Hyesik Kong
- Genomic Research Alliance for Transplantation (GRAfT) and Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Ilker Tunc
- Bioinformatics and Computation Core, NHLBI, Maryland, USA
| | - Komudi Singh
- Bioinformatics and Computation Core, NHLBI, Maryland, USA
| | - Ananth Charya
- Genomic Research Alliance for Transplantation (GRAfT) and Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | | | - Steven Nathan
- Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia, USA
| | - Andrea Cox
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Robert A Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Sean Agbor-Enoh
- Genomic Research Alliance for Transplantation (GRAfT) and Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
31
|
Caicedo A, Zambrano K, Sanon S, Luis Vélez J, Montalvo M, Jara F, Moscoso SA, Vélez P, Maldonado A, Velarde G. The diversity and coexistence of extracellular mitochondria in circulation: A friend or foe of the immune system. Mitochondrion 2021; 58:270-284. [PMID: 33662580 DOI: 10.1016/j.mito.2021.02.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 01/22/2023]
Abstract
The diversity and coexistence of extracellular mitochondria may have a key role in the maintenance of health and progression of disease. Studies report that active mitochondria can be found physiologically outside of cells and circulating in the blood without inducing an inflammatory response. In addition, inactive or harmed mitochondria have been recognized as activators of immune cells, as they play an essential role in diseases characterized by the metabolic deregulation of these cells, such as sepsis. In this review we analyze key aspects regarding the existence of a diversity of extracellular mitochondria, their coexistence in body fluids and their effects on various immune cells. Additionally, we introduce models of how extracellular mitochondria could be interacting to maintain health and affect disease prognosis. Unwrapped mitochondria (freeMitos) can exist as viable, active, inactive or harmed organelles. Mitochondria can also be found wrapped in a membrane (wrappedMitos) that may differ depending on the cell of origin. Mitochondrial fragments can also be present in various body fluids as DAMPs, as mtDNA enclosed in vesicles or as circulating-cell-free mtDNA (ccf-mtDNA). Interestingly, the great quantity of evidence regarding the levels of ccf-mtDNA and their correlation with aging and disease allows for the identification of the diversity, but not type, of extracellular mitochondria. The existence of a diversity of mitochondria and their effects on immune cells opens a new concept in the biomedical field towards the understanding of health, the progression of disease and the development of mitochondria as therapeutic agents.
Collapse
Affiliation(s)
- Andrés Caicedo
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; Sistemas Médicos SIME, Universidad San Francisco de Quito, Quito, Ecuador.
| | - Kevin Zambrano
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands; Instituto de Neurociencias, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Serena Sanon
- Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; Cornell University - Ithaca, United States
| | - Jorge Luis Vélez
- Universidad Central del Ecuador, Facultad de Ciencias Médicas, Quito, Ecuador; Hospital Pablo Arturo Suárez, Unidad de Terapia Intensiva y Centro de Investigación Clínica, Quito, Ecuador
| | - Mario Montalvo
- Hospital Pablo Arturo Suárez, Unidad de Terapia Intensiva y Centro de Investigación Clínica, Quito, Ecuador
| | - Fernando Jara
- Hospital Pablo Arturo Suárez, Unidad de Terapia Intensiva y Centro de Investigación Clínica, Quito, Ecuador
| | - Santiago Aguayo Moscoso
- Hospital Pablo Arturo Suárez, Unidad de Terapia Intensiva y Centro de Investigación Clínica, Quito, Ecuador
| | - Pablo Vélez
- Hospital Pablo Arturo Suárez, Unidad de Terapia Intensiva y Centro de Investigación Clínica, Quito, Ecuador
| | - Augusto Maldonado
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, United States; Hospital General Docente de Calderón, Quito, Ecuador
| | - Gustavo Velarde
- Universidad Central del Ecuador, Facultad de Ciencias Médicas, Quito, Ecuador; Hospital Pablo Arturo Suárez, Unidad de Terapia Intensiva y Centro de Investigación Clínica, Quito, Ecuador
| |
Collapse
|
32
|
Montealegre F, Lyons BM. Fluid Therapy in Dogs and Cats With Sepsis. Front Vet Sci 2021; 8:622127. [PMID: 33718468 PMCID: PMC7947228 DOI: 10.3389/fvets.2021.622127] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/03/2021] [Indexed: 01/20/2023] Open
Abstract
Sepsis is currently defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis may occur secondary to infection anywhere in the body, and its pathogenesis is complex and not yet fully understood. Variations in the host immune response result in diverse clinical manifestations, which complicates clinical recognition and fluid therapy both in humans and veterinary species. Septic shock is a subset of sepsis in which particularly profound circulatory, cellular, and metabolic abnormalities are associated with a greater risk of mortality than with sepsis alone. Although septic shock is a form of distributive shock, septic patients frequently present with hypovolemic and cardiogenic shock as well, further complicating fluid therapy decisions. The goals of this review are to discuss the clinical recognition of sepsis in dogs and cats, the basic mechanisms of its pathogenesis as it affects hemodynamic function, and considerations for fluid therapy. Important pathophysiologic changes, such as cellular interaction, microvascular alterations, damage to the endothelial glycocalyx, hypoalbuminemia, and immune paralysis will be also reviewed. The advantages and disadvantages of treatment with crystalloids, natural and synthetic colloids, and blood products will be discussed. Current recommendations for evaluating fluid responsiveness and the timing of vasopressor therapy will also be considered. Where available, the veterinary literature will be used to guide recommendations.
Collapse
Affiliation(s)
- Federico Montealegre
- Department of Medical and Scientific Affairs, Nova Biomedical, Waltham, MA, United States
| | - Bridget M Lyons
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, United States
| |
Collapse
|
33
|
Dobson GP, Biros E, Letson HL, Morris JL. Living in a Hostile World: Inflammation, New Drug Development, and Coronavirus. Front Immunol 2021; 11:610131. [PMID: 33552070 PMCID: PMC7862725 DOI: 10.3389/fimmu.2020.610131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
We present a brief history of the immune response and show that Metchnikoff's theory of inflammation and phagocytotic defense was largely ignored in the 20th century. For decades, the immune response was believed to be triggered centrally, until Lafferty and Cunningham proposed the initiating signal came from the tissues. This shift opened the way for Janeway's pattern recognition receptor theory, and Matzinger's danger model. All models failed to appreciate that without inflammation, there can be no immune response. The situation changed in the 1990s when cytokine biology was rapidly advancing, and the immune system's role expanded from host defense, to the maintenance of host health. An inflammatory environment, produced by immune cells themselves, was now recognized as mandatory for their attack, removal and repair functions after an infection or injury. We explore the cellular programs of the immune response, and the role played by cytokines and other mediators to tailor the right response, at the right time. Normally, the immune response is robust, self-limiting and restorative. However, when the antigen load or trauma exceeds the body's internal tolerances, as witnessed in some COVID-19 patients, excessive inflammation can lead to increased sympathetic outflows, cardiac dysfunction, coagulopathy, endothelial and metabolic dysfunction, multiple organ failure and death. Currently, there are few drug therapies to reduce excessive inflammation and immune dysfunction. We have been developing an intravenous (IV) fluid therapy comprising adenosine, lidocaine and Mg2+ (ALM) that confers a survival advantage by preventing excessive inflammation initiated by sepsis, endotoxemia and sterile trauma. The multi-pronged protection appears to be unique and may provide a tool to examine the intersection points in the immune response to infection or injury, and possible ways to prevent secondary tissue damage, such as that reported in patients with COVID-19.
Collapse
Affiliation(s)
- Geoffrey P. Dobson
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia
| | | | | | | |
Collapse
|
34
|
Abstract
Traumatic injuries are a leading cause of death and disability in both military and civilian populations. Given the complexity and diversity of traumatic injuries, novel and individualized treatment strategies are required to optimize outcomes. Cellular therapies have potential benefit for the treatment of acute or chronic injuries, and various cell-based pharmaceuticals are currently being tested in preclinical studies or in clinical trials. Cellular therapeutics may have the ability to complement existing therapies, especially in restoring organ function lost due to tissue disruption, prolonged hypoxia or inflammatory damage. In this article we highlight the current status and discuss future directions of cellular therapies for the treatment of traumatic injury. Both published research and ongoing clinical trials are discussed here.
Collapse
|
35
|
Klegeris A. Regulation of neuroimmune processes by damage- and resolution-associated molecular patterns. Neural Regen Res 2021; 16:423-429. [PMID: 32985460 PMCID: PMC7996015 DOI: 10.4103/1673-5374.293134] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Sterile inflammatory processes are essential for the maintenance of central nervous system homeostasis, but they also contribute to various neurological disorders, including neurotrauma, stroke, and demyelinating or neurodegenerative diseases. Immune mechanisms in the central nervous system and periphery are regulated by a diverse group of endogenous proteins, which can be broadly divided into the pro-inflammatory damage-associated molecular patterns (DAMPs) and anti-inflammatory resolution-associated molecular patterns (RAMPs), even though there is notable overlap between the DAMP- and RAMP-like activities for some of these molecules. Both groups of molecular patterns were initially described in peripheral immune processes and pathologies; however, it is now evident that at least some, if not all, of these immunomodulators also regulate neuroimmune processes and contribute to neuroinflammation in diverse central nervous system disorders. The review of recent literature demonstrates that studies on DAMPs and RAMPs of the central nervous system still lag behind the much broader research effort focused on their peripheral counterparts. Nevertheless, this review also reveals that over the last five years, significant advances have been made in our understanding of the neuroimmune functions of several well-established DAMPs, including high-mobility group box 1 protein and interleukin 33. Novel neuroimmune functions have been demonstrated for other DAMPs that previously were considered almost exclusively as peripheral immune regulators; they include mitochondrial transcription factor A and cytochrome C. RAMPs of the central nervous system are an emerging area of neuroimmunology with very high translational potential since some of these molecules have already been used in preclinical and clinical studies as candidate therapeutic agents for inflammatory conditions, such as multiple sclerosis and rheumatoid arthritis. The therapeutic potential of DAMP antagonists and neutralizing antibodies in central nervous system neuroinflammatory diseases is also supported by several of the identified studies. It can be concluded that further studies of DAMPs and RAMPs of the central nervous system will continue to be an important and productive field of neuroimmunology.
Collapse
Affiliation(s)
- Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, Canada
| |
Collapse
|
36
|
XU Z, YIN N, REN R, RUAN Z. In silico analysis based on constituents of the medicinal plant Xuebijing (XBJ) to identify candidate treatment agents for sepsis in the omics-driven research era. MINERVA BIOTECNOL 2021. [DOI: 10.23736/s1120-4826.20.02684-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
37
|
Lv SJ, Zhang GH, Xia JM, Yu H, Zhao F. Early use of high-dose vitamin C is beneficial in treatment of sepsis. Ir J Med Sci 2020; 190:1183-1188. [PMID: 33094466 DOI: 10.1007/s11845-020-02394-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/07/2020] [Indexed: 01/08/2023]
Abstract
PURPOSE Vitamin C has shown benefits in patients with sepsis in addition to standard therapy recently. However, further evidence is required to verify the efficacy of vitamin C in clinical practice. This study aimed to investigate the effect of adjunctive intravenous high-dose vitamin C treatment on hospital mortality in patients with sepsis. METHODS One hundred seventeen patients with sepsis in our department from June 2017 to May 2019 were randomly divided into two groups: the control group (56 cases) and the vitamin C group (61 cases). The control group was treated by the routine and basic therapy with intravenous drip of 5% dextrose and placebo (100 ml/time, 2 times/day), while the vitamin C group was administered intravenously by 3.0 g vitamin C dissolved into 5% dextrose (100 ml/time, 2 times/day) based on the control group. The mortality and efficacy were statistically analyzed and compared between the two groups. RESULTS The 28-day mortality differed significantly between the control group and the vitamin C group (42.97% vs. 27.93%) (p < 0.05). The changes in the sepsis-related organ failure assessment (ΔSOFA) scores at 72 h after ICU admission (4.2 vs. 2.1), the application time of vasoactive drugs (25.6 vs. 43.8), and the procalcitonin clearance (79.6% vs. 61.3%) differed significantly between groups (p < 0.05). CONCLUSION The early treatment of sepsis with intravenous high-dose vitamin C in combination with standard therapy showed a beneficial effect on sepsis, in terms of the reduced 28-day mortality, the decreased SOFA score, and the increased clearance rate of procalcitonin.
Collapse
Affiliation(s)
- Shi-Jin Lv
- Department of Emergency, Hangzhou Normal University Affiliated Hospital, Wenzhou Road 126, Hangzhou, 310015, Zhejiang, People's Republic of China
| | - Guo-Hu Zhang
- Department of Emergency, Hangzhou Normal University Affiliated Hospital, Wenzhou Road 126, Hangzhou, 310015, Zhejiang, People's Republic of China.
| | - Jin-Ming Xia
- Department of Emergency, Hangzhou Normal University Affiliated Hospital, Wenzhou Road 126, Hangzhou, 310015, Zhejiang, People's Republic of China
| | - Huan Yu
- Department of Emergency, Hangzhou Normal University Affiliated Hospital, Wenzhou Road 126, Hangzhou, 310015, Zhejiang, People's Republic of China
| | - Fei Zhao
- Department of Emergency, Hangzhou Normal University Affiliated Hospital, Wenzhou Road 126, Hangzhou, 310015, Zhejiang, People's Republic of China
| |
Collapse
|
38
|
Dobson GP. Trauma of major surgery: A global problem that is not going away. Int J Surg 2020; 81:47-54. [PMID: 32738546 PMCID: PMC7388795 DOI: 10.1016/j.ijsu.2020.07.017] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/27/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022]
Abstract
Globally, a staggering 310 million major surgeries are performed each year; around 40 to 50 million in USA and 20 million in Europe. It is estimated that 1-4% of these patients will die, up to 15% will have serious postoperative morbidity, and 5-15% will be readmitted within 30 days. An annual global mortality of around 8 million patients places major surgery comparable with the leading causes of death from cardiovascular disease and stroke, cancer and injury. If surgical complications were classified as a pandemic, like HIV/AIDS or coronavirus (COVID-19), developed countries would work together and devise an immediate action plan and allocate resources to address it. Seeking to reduce preventable deaths and post-surgical complications would save billions of dollars in healthcare costs. Part of the global problem resides in differences in institutional practice patterns in high- and low-income countries, and part from a lack of effective perioperative drug therapies to protect the patient from surgical stress. We briefly review the history of surgical stress and provide a path forward from a systems-based approach. Key to progress is recognizing that the anesthetized brain is still physiologically 'awake' and responsive to the sterile stressors of surgery. New intravenous drug therapies are urgently required after anesthesia and before the first incision to prevent the brain from switching to sympathetic overdrive and activating secondary injury progression such as hyperinflammation, coagulopathy, immune activation and metabolic dysfunction. A systems-based approach targeting central nervous system-mitochondrial coupling may help drive research to improve outcomes following major surgery in civilian and military medicine.
Collapse
Affiliation(s)
- Geoffrey P Dobson
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland, 4811, Australia.
| |
Collapse
|
39
|
Wollborn J, Steiger C, Ruetten E, Benk C, Kari FA, Wunder C, Meinel L, Buerkle H, Schick MA, Goebel U. Carbon monoxide improves haemodynamics during extracorporeal resuscitation in pigs. Cardiovasc Res 2020; 116:158-170. [PMID: 30873524 DOI: 10.1093/cvr/cvz075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/25/2019] [Accepted: 03/13/2019] [Indexed: 01/26/2023] Open
Abstract
AIMS Heart disease of different aetiology remains the leading cause of cardiac arrest (CA). Despite efforts to improve the quality of cardiopulmonary resuscitation (CPR), subsequent myocardial and systemic damage after CA still present a major long-term burden. Low-dose carbon monoxide (CO) is known to exert protective effects in cardiovascular pathophysiology but clinical applications are challenged by unfavourable delivery modes. We tested the hypothesis that extracorporeal resuscitation (E-CPR) in combination with controlled fast onset CO delivery results in improved cardiac physiology and haemodynamics. Damage-associated molecular pattern (DAMP) signalling may be part of the molecular mechanism. METHODS AND RESULTS In an established porcine model, E-CPR was performed. While E-CPR leads to similar results as compared to a conventional CPR strategy, CO delivery in combination with E-CPR demonstrated significant cardioprotection. Cardiac performance analysis using echocardiography and thermodilution techniques showed a CO-dependent improved cardiac function compared to severe myocardial dysfunction in CPR and E-CPR (left ventricular ejection fraction: Sham 49 ± 5; CPR 26 ± 2; E-CPR 25 ± 2; CO-E-CPR 31 ± 4; P < 0.05). While sublingual microcirculation was significantly compromised in CPR and E-CPR, CO delivery demonstrated a significant improvement in microvascular function (microvascular flow index: Sham 2.9 ± 0.1; CPR 2.2 ± 0.1; E-CPR 1.8 ± 0.1; CO-E-CPR 2.7 ± 0.1; P < 0.01). Histological and serological myocardial damage markers were significantly reduced (hsTroponin-T Sham 0.01 ± 0.001; CPR 1.9 ± 0.2; E-CPR 3.5 ± 1.2; CO-E-CPR 0.5 ± 0.2 ng/mL; P < 0.05). DAMP signalling was decreased ipse facto leading to influence of cardioprotective heat shock and cyclooxygenase response. CONCLUSIONS CO treatment restores myocardial function and improves systemic macro- and microhaemodynamics in E-CPR through a reduction in DAMPs.
Collapse
Affiliation(s)
- Jakob Wollborn
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Christoph Steiger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Germany
| | - Eva Ruetten
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Christoph Benk
- Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany.,Department of Cardiothoracic Surgery, Heart Center - University of Freiburg, Freiburg, Germany
| | - Fabian A Kari
- Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany.,Department of Cardiothoracic Surgery, Heart Center - University of Freiburg, Freiburg, Germany
| | - Christian Wunder
- Department of Anesthesiology and Critical Care, Robert-Bosch-Krankenhaus Stuttgart, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Germany
| | - Hartmut Buerkle
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Martin A Schick
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Ulrich Goebel
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| |
Collapse
|
40
|
Relja B, Land WG. Damage-associated molecular patterns in trauma. Eur J Trauma Emerg Surg 2020; 46:751-775. [PMID: 31612270 PMCID: PMC7427761 DOI: 10.1007/s00068-019-01235-w] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022]
Abstract
In 1994, the "danger model" argued that adaptive immune responses are driven rather by molecules released upon tissue damage than by the recognition of "strange" molecules. Thus, an alternative to the "self versus non-self recognition model" has been provided. The model, which suggests that the immune system discriminates dangerous from safe molecules, has established the basis for the future designation of damage-associated molecular patterns (DAMPs), a term that was coined by Walter G. Land, Seong, and Matzinger. The pathological importance of DAMPs is barely somewhere else evident as in the posttraumatic or post-surgical inflammation and regeneration. Since DAMPs have been identified to trigger specific immune responses and inflammation, which is not necessarily detrimental but also regenerative, it still remains difficult to describe their "friend or foe" role in the posttraumatic immunogenicity and healing process. DAMPs can be used as biomarkers to indicate and/or to monitor a disease or injury severity, but they also may serve as clinically applicable parameters for optimized indication of the timing for, i.e., secondary surgeries. While experimental studies allow the detection of these biomarkers on different levels including cellular, tissue, and circulatory milieu, this is not always easily transferable to the human situation. Thus, in this review, we focus on the recent literature dealing with the pathophysiological importance of DAMPs after traumatic injury. Since dysregulated inflammation in traumatized patients always implies disturbed resolution of inflammation, so-called model of suppressing/inhibiting inducible DAMPs (SAMPs) will be very briefly introduced. Thus, an update on this topic in the field of trauma will be provided.
Collapse
Affiliation(s)
- Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany.
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt, Germany.
| | - Walter Gottlieb Land
- Molecular ImmunoRheumatology, INSERM UMR_S1109, Laboratory of Excellence Transplantex, University of Strasbourg, Strasbourg, France
| |
Collapse
|
41
|
Qian LW, Evani SJ, Chen P, Brandenburg KS, Weaver AJ, Fourcaudot AB, Abercrombie JJ, Sebastian EA, Leung KP. Cerium Nitrate Treatment Provides Eschar Stabilization through Reduction in Bioburden, DAMPs, and Inflammatory Cytokines in a Rat Scald Burn Model. J Burn Care Res 2020; 41:576-584. [PMID: 31808807 PMCID: PMC7195553 DOI: 10.1093/jbcr/irz199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In this study, we used a clinically relevant rat scald burn model to determine the treatment effects of cerium nitrate (CN) for stabilizing burn eschars through reduction of damage-associated molecular patterns (DAMPs), inflammatory cytokines, and bioburden. Forty-two male Sprague-Dawley rats were anesthetized before undergoing a scald burn at 99°C for 6 seconds to create a 10% full-thickness burn. The test groups included sham burn, burn with water bathing, and burn with CN bathing. End point parameters included circulating DAMPs, proinflammatory cytokines, tissue myeloperoxidase activity, and quantification of resident flora in burn skin. The high mobility group protein box 1 was found to be elevated in burn animals at postoperative days (POD) 1 and 7. CN significantly alleviated the increase (P < .05 at POD 1 and P < .01 at POD 7). CN also lessened the heightened levels of hyaluronan in burn animals (P < .05 at POD 7). Additionally, CN significantly reduced the burn-induced increases in interleukin-1β, growth-regulated oncogene/keratinocyte chemoattractant, and macrophage inflammatory protein-1α in burn wounds. The anti-inflammatory effect of CN was also demonstrated in its ability to mitigate the upregulated circulatory xanthine oxidase/dehydrogenase and increased tissue neutrophil infiltration in burn animals. Last, CN suppressed postburn proliferation of resident skin microbes, resulting in a significant 2-log reduction by POD 7. In conclusion, these results suggest that CN attenuates the burn-induced DAMPs, tissue inflammatory responses, and regrowth of resident skin flora, all of which collectively could improve the quality of burn eschar when applied at the point of injury in prolonged field care situations.
Collapse
Affiliation(s)
- Li-Wu Qian
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX
| | - Shankar J Evani
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX
| | - Ping Chen
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX
| | - Kenneth S Brandenburg
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX
| | - Alan J Weaver
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX
| | - Andrea B Fourcaudot
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX
| | - Johnathan J Abercrombie
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX
| | - Eliza A Sebastian
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX
| | - Kai P Leung
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX
| |
Collapse
|
42
|
Inflammatory response in trauma patients: are there ways to decrease the inflammatory reaction? Curr Opin Anaesthesiol 2020; 33:253-258. [PMID: 32049884 DOI: 10.1097/aco.0000000000000842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Trauma patients are considered a complex population of patients in emergency medicine and need extensive, specialized therapy. One major part is the prevention and treatment of the inflammatory response, which occurs in patients after severe injury resulting in complications like endotheliopathy. Likely as a consequence, coagulopathy occurs. Sterile inflammation is hard to address, especially because of the lack of a single activator. Moreover, it is a complex composition of factors that lead to a pathologic immune response. Our understanding of these patterns is increasing, but the complete pathophysiologic changes have yet to be investigated. Therefore, there is no specific target to treat inflammatory response in trauma patients at the moment. RECENT FINDINGS There is increasing knowledge of the pathways and mediators that are responsible for the inflammatory response in patients after severe trauma. The endothelial glycocalyx has been identified to be an integral part of these mechanisms. There have been several new therapeutic approaches to diminish the inflammatory response. SUMMARY Our increasing understanding of the immune system have led to new potential therapeutic perspectives. All of these approaches need further research to be validated. As the current therapies are based on empirical strategies and have not changed much over the years, new treatment options would be an important progress.
Collapse
|
43
|
Aghapour M, Remels AHV, Pouwels SD, Bruder D, Hiemstra PS, Cloonan SM, Heijink IH. Mitochondria: at the crossroads of regulating lung epithelial cell function in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2020; 318:L149-L164. [PMID: 31693390 PMCID: PMC6985875 DOI: 10.1152/ajplung.00329.2019] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/04/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
Disturbances in mitochondrial structure and function in lung epithelial cells have been implicated in the pathogenesis of various lung diseases, including chronic obstructive pulmonary disease (COPD). Such disturbances affect not only cellular energy metabolism but also alter a range of indispensable cellular homeostatic functions in which mitochondria are known to be involved. These range from cellular differentiation, cell death pathways, and cellular remodeling to physical barrier function and innate immunity, all of which are known to be impacted by exposure to cigarette smoke and have been linked to COPD pathogenesis. Next to their well-established role as the first physical frontline against external insults, lung epithelial cells are immunologically active. Malfunctioning epithelial cells with defective mitochondria are unable to maintain homeostasis and respond adequately to further stress or injury, which may ultimately shape the phenotype of lung diseases. In this review, we provide a comprehensive overview of the impact of cigarette smoke on the development of mitochondrial dysfunction in the lung epithelium and highlight the consequences for cell function, innate immune responses, epithelial remodeling, and epithelial barrier function in COPD. We also discuss the applicability and potential therapeutic value of recently proposed strategies for the restoration of mitochondrial function in the treatment of COPD.
Collapse
Affiliation(s)
- Mahyar Aghapour
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control, and Prevention, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany and Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Alexander H V Remels
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Simon D Pouwels
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control, and Prevention, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany and Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Stanford I, Weill Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| |
Collapse
|
44
|
Persistently increased cell-free DNA concentrations only modestly contribute to outcome and host response in sepsis survivors with chronic critical illness. Surgery 2019; 167:646-652. [PMID: 31898953 DOI: 10.1016/j.surg.2019.11.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/14/2019] [Accepted: 11/25/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Although early survival from sepsis has improved with timely resuscitation and source control, survivors frequently experience persistent inflammation and develop chronic critical illness. We examined whether increased copy number of endogenous alarmins, mitochondrial DNA, and nuclear DNA are associated with the early "genomic storm" in blood leukocytes and the development of chronic critical illness in hospitalized patients with surgical sepsis. METHODS A prospective, observational, cohort study of critically ill septic patients was performed at a United States tertiary health care center. Blood samples were obtained at multiple time points after the onset of sepsis. Droplet Digital polymerase chain reaction (Bio-Rad Laboratories, Hercules, CA) was performed to quantify RHO (nuclear DNA) and MT-CO2 (mitochondrial DNA) copies in plasma. Leukocyte transcriptomic expression of 63 genes was also measured in whole blood. RESULTS We enrolled 112 patients with surgical sepsis. Two experienced early death, 69 recovered rapidly, and 41 developed chronic critical illness. Both mitochondrial DNA and nuclear DNA copy number were increased in all sepsis survivors, but early nuclear DNA, and not mitochondrial DNA, copy number was further increased in patients who developed chronic critical illness. Cell-free DNA copy number was associated with in-hospital but not long-term (180-day and 365-day) mortality and were only weakly correlated with leukocyte transcriptomics. CONCLUSION Increased cell-free DNA copy number persists in survivors of sepsis but is not strongly associated with leukocyte transcriptomics. Nuclear DNA but not mitochondrial DNA copy number is associated with adverse, short-term, clinical trajectories and outcomes.
Collapse
|
45
|
Hill BG, Shiva S, Ballinger S, Zhang J, Darley-Usmar VM. Bioenergetics and translational metabolism: implications for genetics, physiology and precision medicine. Biol Chem 2019; 401:3-29. [PMID: 31815377 PMCID: PMC6944318 DOI: 10.1515/hsz-2019-0268] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 06/24/2019] [Indexed: 12/25/2022]
Abstract
It is now becoming clear that human metabolism is extremely plastic and varies substantially between healthy individuals. Understanding the biochemistry that underlies this physiology will enable personalized clinical interventions related to metabolism. Mitochondrial quality control and the detailed mechanisms of mitochondrial energy generation are central to understanding susceptibility to pathologies associated with aging including cancer, cardiac and neurodegenerative diseases. A precision medicine approach is also needed to evaluate the impact of exercise or caloric restriction on health. In this review, we discuss how technical advances in assessing mitochondrial genetics, cellular bioenergetics and metabolomics offer new insights into developing metabolism-based clinical tests and metabolotherapies. We discuss informatics approaches, which can define the bioenergetic-metabolite interactome and how this can help define healthy energetics. We propose that a personalized medicine approach that integrates metabolism and bioenergetics with physiologic parameters is central for understanding the pathophysiology of diseases with a metabolic etiology. New approaches that measure energetics and metabolomics from cells isolated from human blood or tissues can be of diagnostic and prognostic value to precision medicine. This is particularly significant with the development of new metabolotherapies, such as mitochondrial transplantation, which could help treat complex metabolic diseases.
Collapse
Affiliation(s)
- Bradford G. Hill
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, KY 40202
| | - Sruti Shiva
- Department of Pharmacology & Chemical Biology, Vascular Medicine Institute, Center for Metabolism & Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA 15143
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Veteran Affairs Medical Center, Birmingham, AL 35294
| | - Victor M. Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
46
|
Club cell protein 16 in sera from trauma patients modulates neutrophil migration and functionality via CXCR1 and CXCR2. Mol Med 2019; 25:45. [PMID: 31666007 PMCID: PMC6822417 DOI: 10.1186/s10020-019-0115-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 10/16/2019] [Indexed: 01/21/2023] Open
Abstract
Background Club Cell protein (CC)16 correlates with lung injury and respiratory complications, which are in part triggered by polymorphonuclear leukocytes (PMNL) in severely traumatized patients (TP). CC16 exerts anti-inflammatory and immunosuppressive effects, however, its influence on PMNL functions after trauma is unknown. Here, we evaluated whether CC16 present in sera from TP could modify the biological functions of PMNL. Methods Sera from 16 severely injured TP without pneumonia (no P, n = 8) or with pneumonia (P, n = 8) were collected at admission to emergency department (ED) and 1 day prior pneumonia and pre-incubated with or without anti-CC16 antibody for CC16 neutralization. Samples from the equal post-injury days in the corresponding no P group were used. Neutrophils were isolated from healthy volunteers (HV, n = 5) and incubated with 20% of the serum medium from TP, respectively. In PMNL, CD62L, CD11b/CD18 and CD31 expression, migratory capacity, phagocytosis rate, oxidative burst and apoptosis were investigated. In isolated PMNL, CXCR1 and CXCR2 were neutralized before stimulation with CC16, and oxidative burst, phagocytosis and apoptosis were analyzed in neutrophils and their subsets. Results Serum from the P group enhanced significantly PMNL migration compared to no P group, while CC16-neutralization further increased the migratory rate of PMNL in both groups. CC16-neutralization increased significantly the expression of CD62L in the P group at ED. Oxidative burst was significantly increased in the P group vs. no P during the study period. CC16 seemed to have no influence on oxidative burst and phagocytosis in TP. However, in a more controlled study design, CC16 induced a significant increase of oxidative burst and a decrease of apoptosis of CD16+ granulocytes. These effects were markedly observed in mature CD16brightCD62Lbright and immune suppressive CD16brightCD62Ldim neutrophils. In mature subset, CXCR1 and CXCR2 neutralization diminished CC16-induced effects. Conclusions CC16 in sera from multiply traumatized patients, notably of those with pneumonia, has significant effects on PMNL. The results suggest an association of CC16 with CXCR1 and CXCR2. Our data suggest that CC16 reduces the migratory capacity of PMNL and thus modulates their function in patients with respiratory complications after trauma.
Collapse
|
47
|
Stortz JA, Hawkins RB, Holden DC, Raymond SL, Wang Z, Brakenridge SC, Cuschieri J, Moore FA, Maier RV, Moldawer LL, Efron PA. Cell-free nuclear, but not mitochondrial, DNA concentrations correlate with the early host inflammatory response after severe trauma. Sci Rep 2019; 9:13648. [PMID: 31541163 PMCID: PMC6754448 DOI: 10.1038/s41598-019-50044-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/31/2019] [Indexed: 12/23/2022] Open
Abstract
Severe blunt trauma is associated with an early ‘genomic storm’ which causes simultaneous up- and down-regulation of host protective immunity. Excessive inflammation can lead to organ injury. In the absence of infection, the inflammatory response is presumably driven by release of endogenous alarmins called danger-associated molecular patterns (DAMPs), which initiate immune responses through pattern-recognition receptors (PRR). Here we examined the relationship between concentrations of cell-free (cf) nuclear DNA (ncDNA) and mitochondrial DNA (mtDNA) within 24 hours post trauma with circulating leukocyte transcriptomics and plasma IL-6 concentrations, as well as the patients’ clinical trajectories. In 104 patients enrolled from two level-1 trauma centers, ncDNA and mtDNA concentrations were increased within 24 hours of severe trauma, but only ncDNA concentrations correlated with leukocyte gene expression and outcomes. Surprisingly, ncDNA, not mtDNA concentrations, were significantly elevated in trauma patients who developed chronic critical illness versus rapid clinical recovery. Plasma IL-6 and leukocyte transcriptomics were better predictors of outcomes than cfDNA levels. Although mtDNA and ncDNA are significantly increased in the immediate post-trauma period, the dramatic inflammatory and gene expression changes seen after severe trauma are only weakly correlated with ncDNA concentrations, and more importantly, mtDNA concentrations are not associated with adverse clinical trajectories.
Collapse
Affiliation(s)
- Julie A Stortz
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Russell B Hawkins
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - David C Holden
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Steven L Raymond
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Zhongkai Wang
- Department of Biostatistics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Scott C Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Joseph Cuschieri
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Frederick A Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Ronald V Maier
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Philip A Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA.
| |
Collapse
|
48
|
Lee GR, Shaefi S, Otterbein LE. HO-1 and CD39: It Takes Two to Protect the Realm. Front Immunol 2019; 10:1765. [PMID: 31402920 PMCID: PMC6676250 DOI: 10.3389/fimmu.2019.01765] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022] Open
Abstract
Cellular protective mechanisms exist to ensure survival of the cells and are a fundamental feature of all cells that is necessary for adapting to changes in the environment. Indeed, evolution has ensured that each cell is equipped with multiple overlapping families of genes that safeguard against pathogens, injury, stress, and dysfunctional metabolic processes. Two of the better-known enzymatic systems, conserved through all species, include the heme oxygenases (HO-1/HO-2), and the ectonucleotidases (CD39/73). Each of these systems generates critical bioactive products that regulate the cellular response to a stressor. Absence of these molecules results in the cell being extremely predisposed to collapse and, in most cases, results in the death of the cell. Recent reports have begun to link these two metabolic pathways, and what were once exclusively stand-alone are now being found to be intimately interrelated and do so through their innate ability to generate bioactive products including adenosine, carbon monoxide, and bilirubin. These simple small molecules elicit profound cellular physiologic responses that impact a number of innate immune responses, and participate in the regulation of inflammation and tissue repair. Collectively these enzymes are linked not only because of the mitochondria being the source of their substrates, but perhaps more importantly, because of the impact of their products on specific cellular responses. This review will provide a synopsis of the current state of the field regarding how these systems are linked and how they are now being leveraged as therapeutic modalities in the clinic.
Collapse
Affiliation(s)
- Ghee Rye Lee
- Departments of Surgery and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Shahzad Shaefi
- Departments of Surgery and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Leo E Otterbein
- Departments of Surgery and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
49
|
Nedeva C, Menassa J, Puthalakath H. Sepsis: Inflammation Is a Necessary Evil. Front Cell Dev Biol 2019; 7:108. [PMID: 31281814 PMCID: PMC6596337 DOI: 10.3389/fcell.2019.00108] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022] Open
Abstract
Sepsis is one of the leading causes of deaths world-wide and yet there are no therapies available other than ICU treatment. The patient outcome is determined by a complex interplay between the pro and anti-inflammatory responses of the body i.e., a homeostatic balance between these two competing events to be achieved for the patient’s recovery. The initial attempts on drug development mainly focused on controlling inflammation, however, without any tangible outcome. This was despite most deaths occurring during the immune paralysis stage of this biphasic disease. Recently, the focus has been shifting to understand immune paralysis (caused by apoptosis and by anti-inflammatory cytokines) to develop therapeutic drugs. In this review we put forth an argument for a proper understanding of the molecular basis of inflammation as well as apoptosis for developing an effective therapy.
Collapse
Affiliation(s)
- Christina Nedeva
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Joseph Menassa
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Hamsa Puthalakath
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
50
|
|