1
|
Mendes RDS, Silva PL, Robba C, Battaglini D, Lopes-Pacheco M, Caruso-Neves C, Rocco PRM. Advancements in understanding the mechanisms of lung-kidney crosstalk. Intensive Care Med Exp 2024; 12:81. [PMID: 39298036 DOI: 10.1186/s40635-024-00672-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024] Open
Abstract
This narrative review delves into the intricate interplay between the lungs and the kidneys, with a focus on elucidating the pathogenesis of diseases influenced by immunological factors, acid-base regulation, and blood gas disturbances, as well as assessing the effects of various therapeutic modalities on these interactions. Key disorders, such as anti-glomerular basement membrane (anti-GBM) disease, the syndrome of inappropriate antidiuretic hormone secretion (SIADH), and Anti-neutrophil Cytoplasmic Antibodies (ANCA) associated vasculitis (AAV), are also examined to shed light on their underlying mechanisms. This review also explores the relationship between acute respiratory distress syndrome (ARDS) and acute kidney injury (AKI), emphasizing how inflammatory mediators can lead to systemic damage and impact multiple organs. In ARDS, fluid overload exacerbates pulmonary edema, while imbalances in blood volume, such as hypovolemia or hypervolemia, can precipitate renal dysfunction. The review highlights how mechanical ventilation strategies can compromise renal blood flow, trigger systemic inflammation, and induce hemodynamic and neurohormonal alterations, all contributing to lung and kidney damage. The impact of extracorporeal membrane oxygenation (ECMO) on lung-kidney interactions is evaluated, highlighting its role in severe respiratory failure and its renal implications. Emerging therapies, such as mesenchymal stem cells and extracellular vesicles, are discussed as promising avenues to mitigate organ damage and enhance outcomes in critically ill patients. Overall, this review offers a nuanced exploration of lung-kidney dynamics, bridging historical insights with contemporary perspectives. It underscores the clinical significance of these interactions in critically ill patients and advocates for integrated management approaches to optimize patient outcomes.
Collapse
Affiliation(s)
- Renata de Souza Mendes
- Department of Nephrology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Nephrology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Chiara Robba
- IRCCS Policlinico San Martino, Genoa, Italy
- Dipertimento di Scienze Chirurgiche Diagnostiche e Integrate, Policlinico San Martino, IRCCS Per l'Oncologia e Neuroscienze, Università degli Studi di Genova, Genoa, Italy
| | - Denise Battaglini
- IRCCS Policlinico San Martino, Genoa, Italy
- Dipertimento di Scienze Chirurgiche Diagnostiche e Integrate, Policlinico San Martino, IRCCS Per l'Oncologia e Neuroscienze, Università degli Studi di Genova, Genoa, Italy
| | - Miquéias Lopes-Pacheco
- Department of Pediatrics, Center for Cystic Fibrosis and Airway Disease Research, Emory University School of Medicine, Atlanta, GA, USA
| | - Celso Caruso-Neves
- Laboratory of Biochemistry and Cellular Biology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
2
|
Shen SH, Wang RL, Yuan Q, Jian LY, Guo HH, Li HS, Liu XP, Huang RF. The roles of AMPK/mTOR autophagy pathway in the acute kidney injury-induced acute lung injury. CHINESE J PHYSIOL 2023; 66:73-84. [PMID: 37082995 DOI: 10.4103/cjop.cjop-d-22-00122] [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: 04/22/2023] Open
Abstract
Acute kidney injury (AKI) is one of the most challenging clinical problems in kidney disease due to serious complications and high mortality rate, which can lead to acute lung injury (ALI) through inflammatory reactions and oxidative stress. Adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway has been reported to be involved in the development of renal ischemia-reperfusion through autophagy and it remains unclear whether AMPK/mTOR pathway has an effect on the AKI-induced ALI. In this study, we aimed to investigate the effects of autophagy-related AMPK/mTOR signaling pathway on inflammatory factors and oxidative stress in an AKI-induced ALI model. The 48 male Sprague-Dawley rats were divided into four groups randomly: (i) sham, (ii) ischemia/reperfusion injury (IRI), (iii) IRI + rapamycin (RA), and (iv) IRI + 3-methyladenine (3-MA). Unilateral flank incisions were made and right kidneys were excised. The left kidney was subjected to 60 min of ischemia followed by 12, 24, 48, and 72 h of reperfusion. The levels of Scr, blood urea nitrogen (BUN), Wet/Dry ratio, indexes of inflammation, and oxidative stress were assayed. Histological examinations were performed. The protein expression of AMPK, mTOR, LC3-II/LC3-I ratio, and Beclin-1, ULK1 was evaluated by western blotting and immunohistochemistry. Compared to the rats from the sham group, IRI rats showed significantly pulmonary damage after AKI with increased Scr, BUN, Wet/Dry ratio, indexes of inflammation, and oxidative stress. The expression of AMPK, LC3-II/LC3-I ratio, Beclin-1, and ULK1 and were increased, while p62 and mTOR were decreased. In addition, RA treatment significantly attenuated lung injury by promoting autophagy through the activation of the AMPK/mTOR pathway, and 3-MA treatment exhibited adverse effects inversely. Therefore, the activation of the AMPK/mTOR pathway after renal IRI induction could significantly attenuate kidney injury and following AKI-induced ALI by inducing autophagy, which alienates inflammation, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Si-Heng Shen
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Ruo-Lin Wang
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Qi Yuan
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lu-Yong Jian
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Hua-Hui Guo
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - He-Sheng Li
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xue-Pin Liu
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Ren-Fa Huang
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
3
|
Zhang J, Zhou N, Wang Y, Liu T, Cao Y, Feng W, Zheng X. Protective effects of Descurainia sophia seeds extract and its fractions on pulmonary edema by untargeted urine and serum metabolomics strategy. Front Pharmacol 2023; 14:1080962. [PMID: 36865914 PMCID: PMC9971919 DOI: 10.3389/fphar.2023.1080962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Background: Descurainia sophia seeds (DS) is a herbal medicine in traditional Chinese medicine (TCM) for treating lung diseases. We aimed to evaluate the therapeutic effect of DS and five of its fractions upon pulmonary edema (PE) through metabolomics analysis (MA) of urine and serum samples of rats. Methods: A PE model was established by intrathoracic injection of carrageenan. Rats were pretreated with DS extract or its five fractions (polysaccharides (DS-Pol); oligosaccharides (DS-Oli); flavonoid glycosides (DS-FG); flavonoid aglycone (DS-FA); fat oil fraction (DS-FO)) for seven consecutive days. Forty-eight hours after carrageenan injection, lung tissues were subjected to histopathology. MA of urine and serum was done by ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry, respectively. Principal component analysis and orthogonal partial least squares-discriminant analysis were operated for the MA of rats and potential biomarkers related to treatment. Heatmaps and metabolic networks were constructed to explore how DS and its five fractions act against PE. Results: DS and its five fractions could all attenuate pathologic lung injury to different degrees, and DS-Oli, DS-FG, and DS-FO had a more potent effect compared with DS-Pol and DS-FA. DS-Oli, DS-FG, DS-FA, and DS-FO could regulate the metabolic profiles of PE rats, but DS-Pol was less potent. According to MA, the five fractions could improve PE to some degree due to their anti-inflammatory, immunoregulatory, and renoprotective activities by mediating the metabolism of taurine, tryptophan, and arachidonic acid. However, DS-Oli, DS-FG, and DS-FO had more important roles in edema-fluid reabsorption, and reduction of vascular leakage through regulating the metabolism of phenylalanine, sphingolipid and bile acid. Finally, heatmaps and hierarchical clustering analysis indicated DS-Oli, DS-FG, and DS-FO to be more efficacious than DS-Pol or DS-FA against PE. The five fractions of DS had a synergistic effect on PE from different aspects, thereby constituting the entire efficacy of DS. DS-Oli, DS-FG, or DS-FO could be used as an alternative to DS. Conclusion: MA combined with use of DS and its fractions provided novel insights into the mechanism of action of TCM.
Collapse
Affiliation(s)
- Jinying Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ning Zhou
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China,The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Yongxiang Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tong Liu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yumin Cao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China,The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China,Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan, Education Ministry of P.R, Zhengzhou, China,*Correspondence: Weisheng Feng, ; Xiaoke Zheng,
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China,The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China,Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan, Education Ministry of P.R, Zhengzhou, China,*Correspondence: Weisheng Feng, ; Xiaoke Zheng,
| |
Collapse
|
4
|
Mueen RM, Hadi NR. LUNG PROTECTIVE EFFECTS OF CLOPIDOGREL IN POLYMICROBIAL SEPSIS. POLSKI MERKURIUSZ LEKARSKI : ORGAN POLSKIEGO TOWARZYSTWA LEKARSKIEGO 2023; 51:321-329. [PMID: 37756450 DOI: 10.36740/merkur202304104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
OBJECTIVE Aim: The goal of this experiment was to examine if Clopidogrel might protect the lungs during sepsis by modulating the inflammatory and oxidative stress markers. PATIENTS AND METHODS Materials and Methods: Twenty-four adult male Swiss-albino mice aged 8-12 weeks, with a weighing of 20-30 g, were randomized into 4 equal groups (n=6): sham (Laparotomy without cecal ligation and puncture [CLP]), CLP (laparotomy plus CLP), vehicle (DMSO 1 hour prior to CLP), Clopidogrel (50 mg/g IP 1 hour before to CLP). ELISA was used to assess Lung tissue levels of pro-inflammatory and oxidative stress markers. RESULTS Results: F2 isoprostane levels were significantly higher in the sepsis group (p<0.05) in comparison with sham group, while Clopidogrel was considerably lower (p<0.05) in the inflammatory and oxidative stress markers in comparison to sepsis group. Histologically, all mice in the sepsis group had considerable (p=0.05) lung tissue damage, but Clopidogrel considerably decreased lung tissue injury (p=0.05). CONCLUSION Conclusion: Clopidogrel was found to reduce lung tissue cytokine concentrations (IL-1, TNF a, IL-6, F2 isoprostane, GPR 17, MIF) in male mice during CLP-induced polymicrobial sepsis by modulation of pro-inflammatory and oxidative stress cascade signaling pathways, to the best of our abilities, no study has looked at the effect of Clopidogrel on MIF levels.
Collapse
Affiliation(s)
- Ruaa Murtada Mueen
- DEPARTMENT OF PHARMACOLOGY & THERAPEUTICS, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, KUFA, IRAQ
| | - Najah R Hadi
- DEPARTMENT OF PHARMACOLOGY & THERAPEUTICS, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, KUFA, IRAQ
| |
Collapse
|
5
|
Sarkaki A, Hoseinynejad S, Khombi Shooshtari M, Rashno M. Synaptic plasticity and cognitive impairment consequences to acute kidney injury: Protective role of ellagic acid. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:621-628. [PMID: 35911650 PMCID: PMC9282745 DOI: 10.22038/ijbms.2022.62015.13729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 05/17/2022] [Indexed: 11/09/2022]
Abstract
Objectives The goal of the current experiment was to define the efficacy and underlying molecular mechanisms of Ellagic acid (EA) on acute kidney injury (AKI) induced impairment in cognitive and synaptic plasticity in rats. Materials and Methods Administration of 8 ml/kg glycerol (intramuscular) was used to establish the AKI model. Injured animals were treated by EA (25, 50, and 100 mg/kg, daily, gavage) for 14 consecutive days. To approve the renal injuries and the effects of EA on AKI, creatinine values in serum and urea nitrogen (BUN) values in blood were measured. Cognitive performance was investigated using the Morris water maze test. In vivo long-term potentiation (LTP) was recorded from the hippocampus. Then, the level of IL-10β and TNF-α levels were measured using ELISA kits. The integrity index of the Blood-brain barrier (BBB) was assessed by extravasation of Evans blue dye into the brain. Results Glycerol injection increased blood urea nitrogen (BUN) and serum creatinine (Scr) levels significantly in the AKI group, and EA treatment resulted in a significant reduction in BUN levels in all concentration groups. Also, a significant reduction in the cerebral EBD concentrations was demonstrated in EA treatment rats. Moreover, the indexes of brain electrophysiology, spatial learning, and memory were improved in the EA administrated group compared with the AKI rats. Conclusion The current experiment demonstrated the efficacy of EA in hippocampal complication and cognitive dysfunction secondary to AKI via alleviating the inflammation.
Collapse
Affiliation(s)
- Alireza Sarkaki
- Department of Physiology, Faculty of Medicine, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sarkaki Hoseinynejad
- Department of Physiology, Faculty of Medicine, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran, Chronic Renal Failure Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran,Corresponding author: Khojasteh Hoseinynejad. Department of Physiology, Faculty of Medicine, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Maryam Khombi Shooshtari
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Rashno
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran, Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
6
|
COVID-19 impact on the renal system: Pathophysiology and clinical outcomes. Best Pract Res Clin Anaesthesiol 2021; 35:449-459. [PMID: 34511232 PMCID: PMC7920815 DOI: 10.1016/j.bpa.2021.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 02/23/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus disease (COVID-19) causes many deleterious effects throughout the body. Prior studies show that the incidence of acute kidney injury in COVID-19 patients could be as high as 25%. There are also autopsy reports showing evidence of viral tropism to the renal system. In this regard, COVID-19 can damage the kidneys and increase a patient's risk of requiring dialysis. Available evidence suggests that renal involvement in COVID-19 infection is not uncommon, and there has been an increased incidence of chronic kidney disease related to the pandemic. In this literature analysis, we address COVID-19 and its effects on the renal system, including the pathophysiologic mechanisms. We also address current studies on the causes of injury to the renal system, the cause of kidney failure, its effect on mortality, the impact on dialysis patients, and the impact on renal transplant patients. COVID-19 disease may have unique features in individuals on chronic dialysis and kidney transplant recipients, requiring increased vigilance in limiting viral transmission in perioperative, in-patient, and dialysis center settings.
Collapse
|
7
|
Kallet RH. 2020 Year in Review: Mechanical Ventilation During the First Year of the COVID-19 Pandemic. Respir Care 2021; 66:1341-1362. [PMID: 33972456 PMCID: PMC9994377 DOI: 10.4187/respcare.09257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Coronavirus disease 2019 (COVID-19) represents the greatest medical crisis encountered in the young history of critical care and respiratory care. During the early months of the pandemic, when little was known about the virus, the acute hypoxemic respiratory failure it caused did not appear to fit conveniently or consistently into our classification of ARDS. This not only re-ignited a half-century's long simmering debate over taxonomy, but also fueled similar debates over how PEEP and lung-protective ventilation should be titrated, as well as the appropriate role of noninvasive ventilation in ARDS. COVID-19 ignited other debates on emerging concepts such as ARDS phenotypes and patient self-inflicted lung injury from vigorous spontaneous breathing. Over a year later, these early perplexities have receded into the background without having been reviewed or resolved. With a full year of evidence having been published, this narrative review systematically analyzes whether COVID-19-associated respiratory failure is essentially ARDS, with perhaps a somewhat different course of presentation. This includes a review of the severity of hypoxemia and derangements in pulmonary mechanics, PEEP requirements, recruitment potential, ability to achieve lung-protective ventilation goals, duration of mechanical ventilation, associated mortality, and response to noninvasive ventilation. This paper also reviews the concepts of ARDS phenotypes and patient self-inflicted lung injury as these are crucial to understanding the contentious debate over the nature and management of COVID-19.
Collapse
Affiliation(s)
- Richard H Kallet
- Department of Anesthesia and Perioperative Care, University of California, San Francisco at Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California.
| |
Collapse
|
8
|
Delayed hemodialysis in COVID-19: Case series with literature review. Clin Nephrol Case Stud 2021; 9:26-32. [PMID: 33732571 PMCID: PMC7962471 DOI: 10.5414/cncs110240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/09/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Increased incidence of kidney injury has been seen in patients with COVID-19. However, less is known about COVID-19 susceptibility and outcomes in end-stage renal disease (ESRD) patients on hemodialysis (HD). Reduced angiotensin-converting enzyme 2 (ACE-2) from SARS-CoV-2 binding and increased angiotensin II (Ang-II) activity have been suggested as mechanisms for COVID-19 renal pathophysiology. MATERIALS AND METHODS In this case series, we analyzed the data of 3 patients with ESRD who had a delay in receiving their regular HD. Reduced oxygen requirement, resolved hyperkalemia, and normalized fluid status were used for the basis of discharge. RESULTS Presenting symptoms included fever, dyspnea, and dry cough. Laboratory markers were characteristic for COVID-19, such as lymphopenia, elevated D-dimer, C-reactive protein (CRP), and interleukin 6 (IL-6). All 3 of our reported patients required urgent HD upon admission. However, we report no fatalities in our case series, and our patients did not have a severe course of illness requiring endotracheal intubation. We reviewed COVID-19 pathophysiology and how patients with ESRD on HD may be particularly at risk for infection. CONCLUSION New renal failure or ESRD sequelae, such as hyperkalemia, uremic encephalopathy, and fluid overload, can be exacerbated by a delay in receiving HD due to COVID-19 infection. Both direct COVID-19 infection and the challenges this pandemic creates to health care logistics present unique threats to ESRD patients on HD.
Collapse
|
9
|
Bülow Anderberg S, Luther T, Berglund M, Larsson R, Rubertsson S, Lipcsey M, Larsson A, Frithiof R, Hultström M. Increased levels of plasma cytokines and correlations to organ failure and 30-day mortality in critically ill Covid-19 patients. Cytokine 2020; 138:155389. [PMID: 33348065 PMCID: PMC7833204 DOI: 10.1016/j.cyto.2020.155389] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 02/09/2023]
Abstract
Fifteen cytokines were increased at ICU admission in Covid-19 patients. A relatively moderate increase in cytokine concentrations was observed. IL-1ra, IL-6 and IP-10 correlated with respiratory failure and acute kidney injury. IL-8 may be a future biomarker due to its correlation with 30-day mortality.
Background The infection caused by SARS CoV-2 has been postulated to induce a cytokine storm syndrome that results in organ failure and even death in a considerable number of patients. However, the inflammatory response in Corona virus disease-19 (Covid-19) and its potential to cause collateral organ damage has not been fully elucidated to date. This study aims to characterize the acute cytokine response in a cohort of critically ill Covid-19 patients. Method 24 adults with PCR-confirmed Covid-19 were included at time of admission to intensive care a median of eleven days after initial symptoms. Eleven adult patients admitted for elective abdominal surgery with preoperative plasma samples served as controls. All patients were included after informed consent was obtained. 27 cytokines were quantified in plasma. The expression of inflammatory mediators was then related to routine inflammatory markers, SAPS3, SOFA score, organ failure and 30-day mortality. Results A general increase in cytokine expression was observed in all Covid-19 patients. A strong correlation between respiratory failure and IL-1ra, IL-4, IL-6, IL-8 and IP-10 expression was observed. Acute kidney injury development correlated well with increased levels of IL-1ra, IL-6, IL-8, IL-17a, IP-10 and MCP-1. Generally, the cohort demonstrated weaker correlations between cytokine expression and 30-day mortality out of which IL-8 showed the strongest signal in terms of mortality. Conclusion The present study found that respiratory failure, acute kidney injury and 30-day mortality in critically ill Covid-19 patients are associated with moderate increases of a broad range of inflammatory mediators at time of admission.
Collapse
Affiliation(s)
- Sara Bülow Anderberg
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden.
| | - Tomas Luther
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Malin Berglund
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Rolf Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Sten Rubertsson
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Miklos Lipcsey
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden; Hedenstierna Laboratory, Anesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Robert Frithiof
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Michael Hultström
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden; Department of Medical Cell Biology, Integrative Physiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
Hu J, Zhang YM, Miao YF, Zhu L, Yi XL, Chen H, Yang XJ, Wan MH, Tang WF. Effects of Yue-Bi-Tang on water metabolism in severe acute pancreatitis rats with acute lung-kidney injury. World J Gastroenterol 2020; 26:6810-6821. [PMID: 33268963 PMCID: PMC7684462 DOI: 10.3748/wjg.v26.i43.6810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/07/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The complications acute lung injury and acute kidney injury caused by severe inflammation are the main reasons of high mortality of severe acute pancreatitis (SAP). These two complications can both lead to water metabolism and acid-base balance disorders, which could act as additional critical factors affecting the disease trend. Aquaporins (AQPs), which can regulate the transmembrane water transport, have been proved to participate in the pathophysiological process of SAP and the associated complications, such as acute lung injury and acute kidney injury. Thus, exploring herbs that can effectively regulate the expression of AQP in SAP could benefit the prognosis of this disease.
AIM To determine whether Yue-Bi-Tang (YBT) can regulate the water metabolism in rats with severe acute pancreatitis via regulating the expression of aquaporins.
METHODS Sprague-Dawley rats were randomly divided into three groups, sham operation group (SOG), model group (MG), and treatment group (TG). SAP was induced with 3.5% sodium taurocholate in the MG and TG. Rats in the TG were administered with YBT while SOG and MG rats were given the same volume of saline. Blood and tissue samples were harvested to detect serum inflammatory cytokines, histopathological changes, malondialdehyde and superoxide dismutase in the lung, and protein and mRNA expression of kidney injury molecule-1, α-smooth muscle actin, and vimentin in the kidney, and AQP1 and 4 in the lung, pancreas, and kidney.
RESULTS The serum interleukin-10, tumor necrosis factor α, and creatinine levels were higher in the MG than in the SOG. Tumor necrosis factor α level in the TG was lower than that in the MG. Malondialdehyde level in lung tissues was higher than in the SOG. The pathological scores and edema scores of the pancreas, lung, and kidney tissues in the MG were all higher than those in the SOG and TG. The protein expression of AQP4 in lung tissues and AQP1 in kidney tissues in the MG were higher than those in the SOG and TG. The expression of vimentin was significantly higher in the MG than in the SOG. The expression of AQP1 mRNA in the lung and kidney, and AQP4 mRNA in the kidney was up-regulated in the MG compared to the SOG.
CONCLUSION YBT might regulate water metabolism to reduce lung and kidney edema of SAP rats via decreasing AQP expression, and alleviate the tissue inflammatory injury.
Collapse
Affiliation(s)
- Jing Hu
- Department of Integrative Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yu-Mei Zhang
- Department of Traditional Chinese Medicine, Xiang’an Hospital of Xiamen University, Xiamen 361101, Fujian Province, China
| | - Yi-Fan Miao
- Department of Integrative Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Lv Zhu
- Department of Integrative Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xiao-Lin Yi
- Department of Integrative Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Huan Chen
- Department of Integrative Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xi-Jing Yang
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Mei-Hua Wan
- Department of Integrative Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wen-Fu Tang
- Department of Integrative Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
11
|
Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M, Delabranche X, Merdji H, Clere-Jehl R, Schenck M, Fagot Gandet F, Fafi-Kremer S, Castelain V, Schneider F, Grunebaum L, Anglés-Cano E, Sattler L, Mertes PM, Meziani F. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med 2020; 46:1089-1098. [PMID: 32367170 PMCID: PMC7197634 DOI: 10.1007/s00134-020-06062-x] [Citation(s) in RCA: 2000] [Impact Index Per Article: 400.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 04/17/2020] [Indexed: 02/06/2023]
Abstract
Purpose Little evidence of increased thrombotic risk is available in COVID-19 patients. Our purpose was to assess thrombotic risk in severe forms of SARS-CoV-2 infection. Methods All patients referred to 4 intensive care units (ICUs) from two centers of a French tertiary hospital for acute respiratory distress syndrome (ARDS) due to COVID-19 between March 3rd and 31st 2020 were included. Medical history, symptoms, biological data and imaging were prospectively collected. Propensity score matching was performed to analyze the occurrence of thromboembolic events between non-COVID-19 ARDS and COVID-19 ARDS patients. Results 150 COVID-19 patients were included (122 men, median age 63 [53; 71] years, SAPSII 49 [37; 64] points). Sixty-four clinically relevant thrombotic complications were diagnosed in 150 patients, mainly pulmonary embolisms (16.7%). 28/29 patients (96.6%) receiving continuous renal replacement therapy experienced circuit clotting. Three thrombotic occlusions (in 2 patients) of centrifugal pump occurred in 12 patients (8%) supported by ECMO. Most patients (> 95%) had elevated D-dimer and fibrinogen. No patient developed disseminated intravascular coagulation. Von Willebrand (vWF) activity, vWF antigen and FVIII were considerably increased, and 50/57 tested patients (87.7%) had positive lupus anticoagulant. Comparison with non-COVID-19 ARDS patients (n = 145) confirmed that COVID-19 ARDS patients (n = 77) developed significantly more thrombotic complications, mainly pulmonary embolisms (11.7 vs. 2.1%, p < 0.008). Coagulation parameters significantly differed between the two groups. Conclusion Despite anticoagulation, a high number of patients with ARDS secondary to COVID-19 developed life-threatening thrombotic complications. Higher anticoagulation targets than in usual critically ill patients should therefore probably be suggested. Electronic supplementary material The online version of this article (10.1007/s00134-020-06062-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julie Helms
- Service de Médecine Intensive Réanimation, Nouvel Hôpital Civil, Hôpitaux universitaires de Strasbourg, 1, Place de l'Hôpital, 67091, Strasbourg Cedex, France
- ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg (UNISTRA), Strasbourg, France
| | - Charles Tacquard
- Service d'Anesthésie-Réanimation, Nouvel Hôpital Civil, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - François Severac
- Groupe Méthodes en Recherche Clinique (GMRC), Hôpital Civil, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Ian Leonard-Lorant
- Radiology Department, Nouvel Hôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | - Mickaël Ohana
- Radiology Department, Nouvel Hôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | - Xavier Delabranche
- Service d'Anesthésie-Réanimation, Nouvel Hôpital Civil, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Hamid Merdji
- Service de Médecine Intensive Réanimation, Nouvel Hôpital Civil, Hôpitaux universitaires de Strasbourg, 1, Place de l'Hôpital, 67091, Strasbourg Cedex, France
- UMR 1260, Regenerative Nanomedicine (RNM), FMTS, INSERM (French National Institute of Health and Medical Research), Strasbourg, France
| | - Raphaël Clere-Jehl
- Service de Médecine Intensive Réanimation, Nouvel Hôpital Civil, Hôpitaux universitaires de Strasbourg, 1, Place de l'Hôpital, 67091, Strasbourg Cedex, France
- ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg (UNISTRA), Strasbourg, France
| | - Malika Schenck
- Service de Médecine Intensive Réanimation, Hautepierre, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Florence Fagot Gandet
- Service de Médecine Intensive Réanimation, Hautepierre, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Samira Fafi-Kremer
- ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg (UNISTRA), Strasbourg, France
- Laboratoire de Virologie Médicale, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Vincent Castelain
- Service de Médecine Intensive Réanimation, Hautepierre, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Francis Schneider
- Service de Médecine Intensive Réanimation, Hautepierre, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Lélia Grunebaum
- Laboratoire de d'Hématologie, Hautepierre, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Eduardo Anglés-Cano
- Innovative Therapies in Haemostasis, INSERM UMR_S 1140, Université de Paris, 75006, Paris, France
| | - Laurent Sattler
- Laboratoire de d'Hématologie, Hautepierre, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Paul-Michel Mertes
- Service d'Anesthésie-Réanimation, Nouvel Hôpital Civil, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Ferhat Meziani
- Service de Médecine Intensive Réanimation, Nouvel Hôpital Civil, Hôpitaux universitaires de Strasbourg, 1, Place de l'Hôpital, 67091, Strasbourg Cedex, France.
- UMR 1260, Regenerative Nanomedicine (RNM), FMTS, INSERM (French National Institute of Health and Medical Research), Strasbourg, France.
| |
Collapse
|
12
|
Farha N, Munguti C. A Dramatic Presentation of Pulmonary Edema Due to Renal Failure. Kans J Med 2020; 13:56-57. [PMID: 32226582 PMCID: PMC7100944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 11/26/2019] [Indexed: 11/12/2022] Open
Affiliation(s)
- Natalie Farha
- Cleveland Clinic, Internal Medicine Residency Program, Cleveland, OH
| | - Cyrus Munguti
- University of Kansas School of Medicine-Wichita, Department of Internal Medicine, Wichita, KS
| |
Collapse
|
13
|
Ibrahim YF, Fadl RR, Ibrahim S, Gayyed MF, Bayoumi A, Refaie M. Protective effect of febuxostat in sepsis-induced liver and kidney injuries after cecal ligation and puncture with the impact of xanthine oxidase, interleukin 1 β, and c-Jun N-terminal kinases. Hum Exp Toxicol 2020; 39:906-919. [PMID: 32054342 DOI: 10.1177/0960327120905957] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Sepsis is one of the most common causes of death among hospitalized patients. Activity of xanthine oxidase (XO), a reactive oxygen species-producing enzyme, is known to be elevated in septic patients. Our aim was to investigate the possible protective role of XO inhibitor, febuxostat (FEB), in a rat model of sepsis-induced liver and kidney injures. Adult male albino rats were divided into four groups (n = 12 each): sham control, sham + FEB, cecal ligation and puncture (CLP), and CLP + FEB groups. FEB (10 mg/kg per os (p.o.)) was given once daily for 2 days and 30 min prior to laparotomy with CLP. CLP was associated with a high mortality rate accompanied by significant liver and kidney injuries indicated by elevated serum alanine aminotransferase, aspartate aminotransferase, urea, and creatinine levels and confirmed by histopathological tissue injury. Moreover, there was an increase in neutrophil gelatinase-associated lipocalin, uric acid, malondialdehyde, and nitric oxide levels and with decreased superoxide dismutase activity and total antioxidant capacity. In addition, CLP caused increased expression of the inflammatory markers tumor necrosis factor alpha, interleukin 1beta protein levels, and nuclear factor kappa B immunoexpression. Finally, CLP operated rats exhibited an upregulation in the apoptotic mediators, caspase 3, and P-C-Jun N-terminal kinases (JNK) proteins. FEB treatment of CLP rats caused a significant improvement and normalization in all measured parameters. Moreover, FEB amerliorates degenerative histopathological changes and improves the overall survival rate. In conclusion, FEB exhibited a protective effect in sepsis-induced liver and kidney injuries most probably through its anti-inflammatory, antioxidant, and antiapoptotic properties and attenuating JNK signaling pathway secondary to its XO enzyme inhibitory activity.
Collapse
Affiliation(s)
- Y F Ibrahim
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia, Egypt
| | - R R Fadl
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia, Egypt
| | - Sae Ibrahim
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia, Egypt
| | - M F Gayyed
- Department of Pathology, Faculty of Medicine, Minia University, El-Minia, Egypt
| | - Ama Bayoumi
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| | - Mmm Refaie
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia, Egypt
| |
Collapse
|
14
|
Malek M. Brain consequences of acute kidney injury: Focusing on the hippocampus. Kidney Res Clin Pract 2018; 37:315-322. [PMID: 30619687 PMCID: PMC6312775 DOI: 10.23876/j.krcp.18.0056] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/19/2018] [Accepted: 07/31/2018] [Indexed: 12/11/2022] Open
Abstract
The high mortality rates associated with acute kidney injury are mainly due to extra-renal complications that occur following distant-organ involvement. Damage to these organs, which is commonly referred to as multiple organ dysfunction syndrome, has more severe and persistent effects. The brain and its sub-structures, such as the hippocampus, are vulnerable organs that can be adversely affected. Acute kidney injury may be associated with numerous brain and hippocampal complications, as it may alter the permeability of the blood-brain barrier. Although the pathogenesis of acute uremic encephalopathy is poorly understood, some of the underlying mechanisms that may contribute to hippocampal involvement include the release of multiple inflammatory mediators that coincide with hippocampus inflammation and cytotoxicity, neurotransmitter derangement, transcriptional dysregulation, and changes in the expression of apoptotic genes. Impairment of brain function, especially of a structure that has vital activity in learning and memory and is very sensitive to renal ischemic injury, can ultimately lead to cognitive and functional complications in patients with acute kidney injury. The objective of this review was to assess these complications in the brain following acute kidney injury, with a focus on the hippocampus as a critical region for learning and memory.
Collapse
Affiliation(s)
- Maryam Malek
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|