1
|
Molinari L, Rio-Pertuz GD, Priyanka P, Smith A, Maggiore JC, Kennedy J, Gomez H, Seymour CW, Kellum JA. Distribution of Acute and Chronic Kidney Disease Across Clinical Phenotypes for Sepsis. Chest 2024; 166:480-490. [PMID: 38462074 PMCID: PMC11443243 DOI: 10.1016/j.chest.2024.03.006] [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: 08/31/2023] [Revised: 02/27/2024] [Accepted: 03/03/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Sepsis is the most common cause of acute kidney injury (AKI) in critically ill patients. Four phenotypes (α, β, γ, δ) for sepsis, which have different outcomes and responses to treatment, were described using routine clinical data in the electronic health record. RESEARCH QUESTION Do the frequencies of AKI, acute kidney disease (AKD), chronic kidney disease (CKD), and AKI on CKD differ by sepsis phenotype? STUDY DESIGN AND METHODS This was a secondary analysis of a randomized clinical trial of early resuscitation, including patients with septic shock at 31 sites. After excluding patients with end-stage kidney disease and missing data, we determined frequencies of the following clinical outcomes: AKI (defined within 24 h as Kidney Disease: Improving Global Outcomes stages 2 or 3 or stage 1 with tissue inhibitor of metalloproteinases-2 × insulin-like growth factor binding protein 7 value of > 2.0), CKD, and AKD (persistence of AKI at any stage on day 7 after enrollment) across four phenotypes. We performed multivariable logistic regression to assess the risk-adjusted association between development of AKI and AKD and phenotype. RESULTS Among 1,090 eligible patients, 543 patients (50%) had AKI. Across phenotypes, the frequencies of AKI varied, being highest in the δ and β phenotypes (78% and 71%, respectively) and the lowest in the α phenotype (26%; P < .001). AKD occurred most often in the δ phenotype (41%) and least often in the α phenotype (8%; P < .001). The highest frequencies of CKD and of AKI on CKD were found in the β phenotype (53% and 38% respectively; P < .001 for both). In the multivariable logistic regression models (α phenotype as reference), δ phenotype showed the strongest association with AKI (OR, 12.33; 95% CI, 7.81-19.47; P < .001) and AKD (OR, 9.18; 95% CI, 5.44-15.51; P < .001). INTERPRETATION The rates of AKI and AKD differed across clinical sepsis phenotypes and are more common among patients with phenotypes β and δ. Phenotype β showed a higher level of underlying CKD that predisposed patients to new AKI. The α and γ phenotypes showed lower frequencies of AKI and less progression to AKD.
Collapse
Affiliation(s)
- Luca Molinari
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA; Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA; Department of Translational Medicine, Università degli Studi del Piemonte Orientale, Novara, Italy
| | - Gaspar Del Rio-Pertuz
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA; Division of Cardiology, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Priyanka Priyanka
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA
| | - Ali Smith
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Joseph C Maggiore
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA
| | - Jason Kennedy
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Hernando Gomez
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA; Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA
| | | | - John A Kellum
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA; Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA.
| |
Collapse
|
2
|
Wang Y, Cui C, Zhao W, Tian X, Liu P, Wei L, Zhu Z, Liu M, Fu R, Jia L. WIP1-mediated regulation of p38 MAPK signaling attenuates pyroptosis in sepsis-associated acute kidney injury. Immunobiology 2024; 229:152832. [PMID: 38943814 DOI: 10.1016/j.imbio.2024.152832] [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: 01/27/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/01/2024]
Abstract
Wild-Type p53-Induced Phosphatase 1 (WIP1/PPM1D) is a serine/threonine phosphatase that plays a significant role in various physiological processes. However, the involvement of WIP1 in kidney remains unclear. Lipopolysaccharide (LPS) was administered to induce acute injury in mice and human kidney 2 (HK2) cells in the study. The WIP1 inhibitor, CCT007093, was administered both in vitro and in vivo to assess its effect on kidney. The single-cell sequencing (scRNA-seq) data revealed that Ppm1d mRNA reached peak on day 2 following unilateral ischemia-reperfusion injury (uni-IRI) in mice, especially in the proximal renal tubules during repair phase. Compared to the control group, WIP1 protein exhibited a significant increase in renal tubules of patients with acute tubular injury (ATI) and mice with LPS-induced acute kidney injury (AKI), as well as in LPS-injured HK2 cells. In vitro experiments showed that CCT007093 increased the protein levels of NLRP3, cleaved-Caspase1, GSDMD-N and IL-1β in HK2 cells and further reduced the viability of LPS-stimulated HK2 cells. In vivo experiments showed that inhibition of WIP1 activity with CCT007093 further increased cleaved-Caspase1, GSDMD-N protein levels in kidney tissue from mice with LPS-induced AKI. In addition, LPS induces phosphorylation of p38 MAPK, a key regulator of pyroptosis, which is further activated by CCT007093. In conclusion, inhibition of WIP1 activity acts as a positive regulator of renal tubular pyroptosis mainly through the mediation of phospho-p38 MAPK.
Collapse
Affiliation(s)
- Yinhong Wang
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chenkai Cui
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Weihao Zhao
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xuefei Tian
- Section of Nephrology, Department of Internal medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Pengfei Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Linting Wei
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zikun Zhu
- Department of Computer Science, School of Computing & Department of Electrical and Computer Engineering, National University of Singapore, Singapore
| | - Ming Liu
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Rongguo Fu
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Lining Jia
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
3
|
Vermeire B, Walsh M, Cox E, Devriendt B. The lipopolysaccharide structure affects the detoxifying ability of intestinal alkaline phosphatases. BMC Vet Res 2024; 20:358. [PMID: 39127648 DOI: 10.1186/s12917-024-04208-3] [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: 11/23/2022] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Lipopolysaccharide (LPS) is one of the most potent mediators of inflammation. In swine husbandry, weaning is associated with LPS-induced intestinal inflammation, resulting in decreased growth rates due to malabsorption of nutrients by the inflamed gut. A potential strategy to treat LPS-mediated disease is administering intestinal alkaline phosphatase (IAP). The latter can detoxify lipid A, the toxic component of LPS, by removal of phosphate groups. Currently, 183 LPS O-serotypes from E. coli have been described, however, comparative experiments to elucidate functional differences between LPS serotypes are scarce. In addition, these functional differences might affect the efficacy of LPS detoxifying enzymes. Here, we evaluated the ability of four LPS serotypes (O26:B6, O55:B5, O111:B4 and O127:B8) derived from Escherichia coli to trigger the secretion of pro-inflammatory cytokines by porcine PBMCs. We also tested the ability of three commercially available IAPs to detoxify these LPS serotypes. The results show that LPS serotypes differ in their ability to trigger cytokine secretion by immune cells, especially at lower concentrations. Moreover, IAPs displayed a different detoxification efficiency of the tested serotypes. Together, this study sheds light on the impact of LPS structure on the detoxification by IAPs. Further research is however needed to elucidate the LPS serotype-specific effects and their implications for the development of novel treatment options to alleviate LPS-induced gut inflammation in weaned piglets.
Collapse
Affiliation(s)
- Bjarne Vermeire
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, Merelbeke, 9820, Belgium
| | | | - Eric Cox
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, Merelbeke, 9820, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, Merelbeke, 9820, Belgium.
| |
Collapse
|
4
|
Steenvoorden TS, Rood JAJ, Bemelman FJ, Armstrong Jr. R, Leuvenink HGD, van der Heijden JW, Vogt L. Alkaline phosphatase treatment of acute kidney injury-an update. Nephrol Dial Transplant 2024; 39:1239-1247. [PMID: 38400561 PMCID: PMC11334066 DOI: 10.1093/ndt/gfae028] [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: 11/02/2023] [Indexed: 02/25/2024] Open
Abstract
Through improved insights into the increasing incidence and detrimental effects of acute kidney injury (AKI), its clinical relevance has become more and more apparent. Although treatment strategies for AKI have also somewhat improved, an adequate remedy still does not exist. Finding one is complicated by a multifactorial pathophysiology and by heterogeneity in the patient population. Alkaline phosphatase (ALP) has been suggested as a therapy for sepsis-associated AKI because of its protective effects against lipopolysaccharide (LPS)-induced inflammation and kidney injury in animals. However, its effectiveness as an AKI treatment has not been demonstrated definitively. Because the anti-inflammatory properties of ALP are likely not reliant on a direct effect on LPS itself, we postulate that other pathways are much more important in explaining the renoprotective properties ascribed to ALP. The re-evaluation of which properties of the ALP enzyme are responsible for the benefit seen in the lab is an important step in determining where the true potential of ALP as a treatment strategy for AKI in the clinic lies. In this review we will discuss how ALP can prevent activation of harmful pro-inflammatory receptors, redirect cell-cell signalling and protect barrier tissues, which together form the basis for current knowledge of the role of ALP in the kidney. With this knowledge in mind and by analysing currently available clinical evidence, we propose directions for new research that can determine whether ALP as a treatment strategy for AKI has a future in the clinical field.
Collapse
Affiliation(s)
- Thei S Steenvoorden
- Department of Internal Medicine, Section Nephrology, Amsterdam UMC, location AMC, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Janneke A J Rood
- Department of Internal Medicine, Section Nephrology, Amsterdam UMC, location VU University Medical Center, Amsterdam, Vrije Universiteit, Amsterdam, The Netherlands
| | - Frederike J Bemelman
- Department of Internal Medicine, Section Nephrology, Amsterdam UMC, location AMC, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Roberto Armstrong Jr.
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Henri G D Leuvenink
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Liffert Vogt
- Department of Internal Medicine, Section Nephrology, Amsterdam UMC, location AMC, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Iba T, Helms J, Maier CL, Levi M, Scarlatescu E, Levy JH. The role of thromboinflammation in acute kidney injury among patients with septic coagulopathy. J Thromb Haemost 2024; 22:1530-1540. [PMID: 38382739 DOI: 10.1016/j.jtha.2024.02.006] [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: 12/01/2023] [Revised: 01/22/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024]
Abstract
Inflammation and coagulation are critical self-defense mechanisms for mitigating infection that can nonetheless induce tissue injury and organ dysfunction. In severe cases, like sepsis, a dysregulated thromboinflammatory response may result in multiorgan dysfunction. Sepsis-associated acute kidney injury (AKI) is a significant contributor to patient morbidity and mortality. The connection between AKI and thromboinflammation is largely due to unique aspects of the renal vasculature. Specifically, the interaction between blood cells with the endothelial, glomerular, and peritubular capillary systems during thromboinflammation reduces oxygen supply to tubular epithelial cells. Previous studies have focused on tubular epithelial cell damage due to hypoxia, oxidative stress, and nephrotoxins. Although these factors are pivotal in acute tubular injury or necrosis, recent studies have demonstrated that AKI in sepsis encompasses a mixture of tubular and glomerular damage subtypes. In cases of sepsis-induced coagulopathy, thromboinflammation within the glomerulus and peritubular capillaries is an important pathogenic mechanism for AKI. Unfortunately, and despite the use of renal replacement therapy, the development of AKI in sepsis continues to be associated with high morbidity, mortality, and clinical challenges requiring alternative approaches. This review introduces the important role of thromboinflammation in AKI pathogenesis and details innovative vascular-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Julie Helms
- French National Institute of Health and Medical Research, United Medical Resources 1260, Regenerative Nanomedicine, Federation de Medicine Translationnelle de Strasbourg, Strasbourg University Hospital, Medical Intensive Care Unit - NHC, Strasbourg University, Strasbourg, France
| | - Cheryl L Maier
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Marcel Levi
- Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands; Department of Medicine, University College London Hospitals National Health Service Foundation Trust, Cardio-metabolic Programme-National Institute for Health and Care Research University College London Hospitals/University College London Biomedical Research Centre, London, United Kingdom
| | - Ecaterina Scarlatescu
- University of Medicine and Pharmacy "Carol Davila," Bucharest, Romania; Department of Anaesthesia and Intensive Care, Fundeni Clinical Institute, Bucharest, Romania
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
6
|
Zhao Y, Zang B, Wang Q. The effectiveness of alkaline phosphatase in sepsis-associated acute kidney injury. Intensive Care Med 2024; 50:778-780. [PMID: 38563896 DOI: 10.1007/s00134-024-07392-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2024] [Indexed: 04/04/2024]
Affiliation(s)
- Yang Zhao
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bin Zang
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Qian Wang
- Department of Emergency, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
7
|
Wu JJ, Zhang TY, Qi YH, Zhu MY, Fang Y, Qi CJ, Cao LO, Lu JF, Lu BH, Tang LM, Shen JX, Mou S. Efficacy and safety of Yiqi Peiyuan granules for improving the short-term prognosis of patients with acute kidney injury: A multicenter, double-blind, placebo-controlled, randomized trial. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:279-285. [PMID: 38688809 DOI: 10.1016/j.joim.2024.04.004] [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: 11/04/2022] [Accepted: 05/12/2023] [Indexed: 05/02/2024]
Abstract
BACKGROUND Yiqi Peiyuan (YQPY) prescription, a composite prescription of traditional Chinese medicine, has been used to prevent or delay the continued deterioration of renal function after acute kidney injury (AKI) in some institutions and has shown considerable efficacy. OBJECTIVE This is the first randomized controlled trial to assess efficacy and safety of YQPY for improving short-term prognosis in adult patients with AKI. DESIGN, SETTING, PARTICIPANTS AND INTERVENTIONS This is a prospective, double-blind, multicenter, randomized, and placebo-controlled clinical trial. A total of 144 enrolled participants were randomly allocated to two groups according to a randomization schedule. Participants, caregivers and investigators assessing the outcomes were blinded to group assignment. Patients in the YQPY group received 36 g YQPY granules twice a day for 28 days. Patients in the placebo group received a placebo in the same dose as the YQPY granules. MAIN OUTCOME MEASURES The primary outcome was the change in the estimated glomerular filtration rate (eGFR) between baseline and after 4 and 24 weeks of treatment. The secondary outcomes were the change of serum creatinine (Scr) level between baseline and after treatment, and the incidence of endpoint events, defined as eGFR increasing by more than 25% above baseline, eGFR >75 mL/min per 1.73 m2 or the composite endpoint, which was defined as the sum of patients meeting either of the above criteria. RESULTS Data from a total of 114 patients (59 in the YQPY group and 55 in the control group) were analyzed. The mean changes in eGFR and Scr in weeks 4 and 24 had no difference between the two groups. In further subgroup analysis (22 in the YQPY group and 31 in the control group), the mean change in eGFR after treatment for 4 weeks was 27.39 mL/min per 1.73 m2 in the YQPY group and 5.78 mL/min per 1.73 m2 in the placebo group, and the mean difference between groups was 21.61 mL/min per 1.73 m2 (P < 0.001). Thirteen (59.1%) patients in the YQPY group and 5 (16.1%) in the placebo group reached the composite endpoints (P = 0.002). During the intervention, 2 and 4 severe adverse events were reported in the YQPY and placebo groups, respectively. CONCLUSION The YQPY granules can effectively improve the renal function of patients 4 weeks after the onset of AKI, indicating that it has good efficacy for improving short-term renal outcomes in patients with AKI. The YQPY granules may be a promising therapy for adults with AKI. TRIAL REGISTRATION Chinese Clinical Trial Registry ChiCTR2100051723. Please cite this article as: Wu JJ, Zhang TY, Qi YH, Zhu MY, Fang Y, Qi CJ, Cao LO, Lu JF, Lu BH, Tang LM, Shen JX, Mou S. Efficacy and safety of Yiqi Peiyuan granules for improving the short-term prognosis of patients with acute kidney injury: a multicenter, double-blind, placebo-controlled, randomized trial. J Integr Med. 2024; 22(3): 279-285.
Collapse
Affiliation(s)
- Jia-Jia Wu
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Uremia Diagnosis and Treatment Center, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Tian-Yi Zhang
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Uremia Diagnosis and Treatment Center, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Ying-Hui Qi
- Department of Nephrology, Shanghai Pudong New Area Punan Hospital, Shanghai 200125, China
| | - Min-Yan Zhu
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Uremia Diagnosis and Treatment Center, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Yan Fang
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Uremia Diagnosis and Treatment Center, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Chao-Jun Qi
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Uremia Diagnosis and Treatment Center, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Li-Ou Cao
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Uremia Diagnosis and Treatment Center, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Ji-Fang Lu
- Department of Nephrology, Ningbo Hangzhou Bay Hospital, Ningbo 315336, Zhejiang Province, China
| | - Bo-Han Lu
- Department of Nephrology, Ningbo Hangzhou Bay Hospital, Ningbo 315336, Zhejiang Province, China
| | - Lu-Min Tang
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Uremia Diagnosis and Treatment Center, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China.
| | - Jian-Xiao Shen
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Uremia Diagnosis and Treatment Center, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China.
| | - Shan Mou
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Uremia Diagnosis and Treatment Center, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
8
|
Pijpe A, Papendorp SG, van der Heijden JW, Vermin B, Ertugrul I, Ritt MWJ, Stessel B, Callebaut I, Beishuizen A, Vlig M, Jimmink J, Huijgen HJ, van Zuijlen PPM, Middelkoop E, de Jong E. Efficacy of Alkaline Phosphatase in Critically Ill Patients with COVID-19: A Multicentre Investigator-Initiated Double-Blind Randomised Placebo-Controlled Trial. Biomedicines 2024; 12:723. [PMID: 38672081 PMCID: PMC11048668 DOI: 10.3390/biomedicines12040723] [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: 02/26/2024] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Efforts to identify therapies to treat hospitalised patients with COVID-19 are being continued. Alkaline phosphatase (AP) dephosphorylates pro-inflammatory adenosine triphosphate (ATP) into anti-inflammatory adenosine. METHODS In a randomised controlled trial, we investigated the safety and efficacy of AP in patients with SARS-CoV-2 infection admitted to the ICU. AP or a placebo was administered for four days following admission to the ICU. The primary outcome was the duration of mechanical ventilation. Mortality in 28 days, acute kidney injury, need for reintubation, safety, and inflammatory markers relevant to the described high cytokine release associated with SARS-CoV-2 infection were the secondary outcomes. RESULTS Between December 2020 and March 2022, 97 patients (of the intended 132) were included, of which 51 were randomised to AP. The trial was terminated prematurely based on meeting the threshold for futility. Compared to the placebo, AP did not affect the duration of mechanical ventilation (9.0 days vs. 9.3 days, p = 1.0). No safety issues were observed. After 28 days, mortality was 9 (18%) in the AP group versus 6 (13%) in the placebo group (p = 0.531). Additionally, no statistically significant differences between the AP and the placebo were observed for the other secondary outcomes. CONCLUSIONS Alkaline phosphatase (AP) therapy in COVID-19 ICU patients showed no significant benefits in this trial.
Collapse
Affiliation(s)
- Anouk Pijpe
- Department of Intensive Care, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands; (A.P.); (S.G.P.)
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC Location Vrije Universiteit, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (P.P.M.v.Z.); (E.M.)
- Amsterdam Movement Sciences, Tissue Function and Regeneration, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Association of Dutch Burn Centres, Zeestraat 27-29, 1941 AJ Beverwijk, The Netherlands;
- Burn Centre, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands;
| | - Stephan G. Papendorp
- Department of Intensive Care, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands; (A.P.); (S.G.P.)
- Burn Centre, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands;
| | - Joost W. van der Heijden
- Department of Internal Medicine, Spaarne Gasthuis, Spaarnepoort 1, 2134 TM Hoofddorp, The Netherlands;
| | - Ben Vermin
- Department of Intensive Care Medicine, Spaarne Gasthuis, Spaarnepoort 1, 2134 TM Hoofddorp, The Netherlands; (B.V.)
| | - Iris Ertugrul
- Department of Intensive Care Medicine, Spaarne Gasthuis, Spaarnepoort 1, 2134 TM Hoofddorp, The Netherlands; (B.V.)
| | - Michael W. J. Ritt
- Department of Intensive Care, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands; (A.P.); (S.G.P.)
| | - Björn Stessel
- Department of Intensive Care Medicine, Jessa Hospital, Stadsomvaart 11, 3500 Hasselt, Belgium; (B.S.); (I.C.)
- LCRC, Faculty of Medicine and Life Sciences, University Hasselt, Agoralaan, 3590 Diepenbeek, Belgium
| | - Ina Callebaut
- Department of Intensive Care Medicine, Jessa Hospital, Stadsomvaart 11, 3500 Hasselt, Belgium; (B.S.); (I.C.)
- LCRC, Faculty of Medicine and Life Sciences, University Hasselt, Agoralaan, 3590 Diepenbeek, Belgium
| | - Albertus Beishuizen
- Intensive Care Center, Medisch Spectrum Twente, Koningsplein 1, 7512 KZ Enschede, The Netherlands;
| | - Marcel Vlig
- Association of Dutch Burn Centres, Zeestraat 27-29, 1941 AJ Beverwijk, The Netherlands;
| | - Joost Jimmink
- Burn Centre, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands;
| | - Henk J. Huijgen
- Department of Clinical Chemistry, Red Cross Hospital, 1942 LE Beverwijk, The Netherlands;
| | - Paul P. M. van Zuijlen
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC Location Vrije Universiteit, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (P.P.M.v.Z.); (E.M.)
- Amsterdam Movement Sciences, Tissue Function and Regeneration, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Burn Centre, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands;
- Department of Plastic Reconstructive and Hand Surgery, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands
- Emma Children’s Hospital, Pediatric Surgical Center, Amsterdam UMC Location Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Esther Middelkoop
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC Location Vrije Universiteit, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (P.P.M.v.Z.); (E.M.)
- Amsterdam Movement Sciences, Tissue Function and Regeneration, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Association of Dutch Burn Centres, Zeestraat 27-29, 1941 AJ Beverwijk, The Netherlands;
- Burn Centre, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands;
| | - Evelien de Jong
- Department of Intensive Care, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands; (A.P.); (S.G.P.)
- Burn Centre, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands;
| |
Collapse
|
9
|
Costigan C, Balgobin S, Zappitelli M. Drugs in treating paediatric acute kidney injury. Pediatr Nephrol 2023; 38:3923-3936. [PMID: 37052689 DOI: 10.1007/s00467-023-05956-4] [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: 01/03/2023] [Revised: 03/03/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023]
Abstract
Acute kidney injury (AKI) is a complex syndrome which affects a significant proportion of hospitalized children. The breadth and impact of AKI on health outcomes in both adults and children have come to the fore in recent years with increasing awareness encouraging research advancement. Despite this, management strategies for most types of AKI remain heavily reliant on fluid and electrolyte management, hemodynamic optimization, nephrotoxin avoidance and appropriate initiation of kidney replacement therapy. Specific drugs targeting the mechanisms involved in AKI remain elusive. Recent improvement in appreciation of the complexity of AKI pathophysiology has allowed for greater opportunity to consider novel therapeutic agents. A number of drugs specifically targeting AKI are in various stages of development. This review will consider some novel and repurposed agents; interrogate the plausibility of the proposed mechanisms of action, as they relate to what we know about the pathophysiology of AKI; and review the level of existing literature supporting their efficacy. The evidence base, particularly in children, is limited.
Collapse
Affiliation(s)
- Caoimhe Costigan
- Department of Pediatrics, Division of Nephrology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Steve Balgobin
- Department of Pediatrics, Division of Nephrology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Michael Zappitelli
- Department of Pediatrics, Division of Nephrology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
- Peter Gilgan Centre for Research and Learning, 686 Bay Street, 11th floor, Rm 11.9722, Toronto, ON, M5G 0A4, Canada.
| |
Collapse
|
10
|
Leite RO, de Souza PO, Haas CB, da Silveira F, Mohr KR, Bertoni APS, Soares MS, Azambuja JH, Prá MD, da Cruz LLP, Gelsleichter NE, Begnini K, Hasko G, Wink MR, Spanevello RM, Braganhol E. ATPergic signaling disruption in human sepsis as a potential source of biomarkers for clinical use. Clin Exp Med 2023; 23:3651-3662. [PMID: 36943594 PMCID: PMC10511658 DOI: 10.1007/s10238-023-01045-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023]
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated inflammatory response to infection. To date, there is no specific treatment established for sepsis. In the extracellular compartment, purines such as adenosine triphosphate (ATP) and adenosine play essential roles in the immune/inflammatory responses during sepsis and septic shock. The balance of extracellular levels among ATP and adenosine is intimately involved in the signals related to immune stimulation/immunosuppression balance. Specialized enzymes, including CD39, CD73, and adenosine deaminase (ADA), are responsible to metabolize ATP to adenosine which will further sensitize the P2 and P1 purinoceptors, respectively. Disruption of the purinergic pathway had been described in the sepsis pathophysiology. Although purinergic signaling has been suggested as a potential target for sepsis treatment, the majority of data available were obtained using pre-clinical approaches. We hypothesized that, as a reflection of deregulation on purinergic signaling, septic patients exhibit differential measurements of serum, neutrophils and monocytes purinergic pathway markers when compared to two types of controls (healthy and ward). It was observed that ATP and ADP serum levels were increased in septic patients, as well as the A2a mRNA expression in neutrophils and monocytes. Both ATPase/ADPase activities were increased during sepsis. Serum ATP and ADP levels, and both ATPase and ADPase activities were associated with the diagnosis of sepsis, representing potential biomarkers candidates. In conclusion, our results advance the translation of purinergic signaling from pre-clinical models into the clinical setting opening opportunities for so much needed new strategies for sepsis and septic shock diagnostics and treatment.
Collapse
Affiliation(s)
- R O Leite
- Departamento de Clínica Médica, Universidade Federal de Pelotas (UFPel), Pelotas, Brazil
- Hospital Nossa Senhora da Conceição, Porto Alegre, Brazil
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, UFPel, Pelotas, Brazil
| | - P O de Souza
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - C B Haas
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - F da Silveira
- Hospital Nossa Senhora da Conceição, Porto Alegre, Brazil
| | - K R Mohr
- Hospital Nossa Senhora da Conceição, Porto Alegre, Brazil
| | - A P S Bertoni
- Programa de Pós-Graduação em Ciências da Saúde, UFCSPA, Porto Alegre, Brazil
| | - M S Soares
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, UFPel, Pelotas, Brazil
| | - J H Azambuja
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - M Dal Prá
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - L L P da Cruz
- Programa de Pós-Graduação em Ciências da Saúde, UFCSPA, Porto Alegre, Brazil
| | - N E Gelsleichter
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - K Begnini
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - G Hasko
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - M R Wink
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, UFCSPA, Porto Alegre, Brazil
| | - R M Spanevello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, UFPel, Pelotas, Brazil
| | - E Braganhol
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, UFPel, Pelotas, Brazil.
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil.
- Programa de Pós-Graduação em Ciências da Saúde, UFCSPA, Porto Alegre, Brazil.
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Sarmento Leite St, 245-Main Building-Room 304, Porto Alegre, RS, 90.050-170, Brazil.
| |
Collapse
|
11
|
Kahrovic A, Poschner T, Schober A, Angleitner P, Alajbegovic L, Andreas M, Hutschala D, Brands R, Laufer G, Wiedemann D. Increased Drop in Activity of Alkaline Phosphatase in Plasma from Patients with Endocarditis. Int J Mol Sci 2023; 24:11728. [PMID: 37511497 PMCID: PMC10380209 DOI: 10.3390/ijms241411728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
(1) Infective endocarditis is a severe inflammatory disease associated with substantial mortality and morbidity. Alkaline phosphatase (AP) levels have been shown to change significantly during sepsis. Additionally, we previously found that a higher initial AP drop after cardiac surgery is associated with unfavorable outcomes. Therefore, the course of AP after surgery for endocarditis is of special interest. (2) A total of 314 patients with active isolated left-sided infective endocarditis at the Department of Cardiac Surgery (Medical University of Vienna, Vienna, Austria) between 2009 and 2018 were enrolled in this retrospective analysis. Blood samples were analyzed at different time points (baseline, postoperative days 1-7, postoperative days 14 and 30). Patients were categorized according to relative alkaline phosphatase drop (≥30% vs. <30%). (3) A higher rate of postoperative renal replacement therapy with or without prior renal replacement therapy (7.4 vs. 21.8%; p = 0.001 and 6.7 vs. 15.6%; p = 0.015, respectively) and extracorporeal membrane oxygenation (2.2 vs. 19.0%; p = 0.000) was observed after a higher initial alkaline phosphatase drop. Short-term (30-day mortality 3.0 vs. 10.6%; p = 0.010) and long-term mortality (p = 0.008) were significantly impaired after a higher initial alkaline phosphatase drop. (4) The higher initial alkaline phosphatase drop was accompanied by impaired short- and long-term outcomes after cardiac surgery for endocarditis. Future risk assessment scores for cardiac surgery should consider alkaline phosphatase.
Collapse
Affiliation(s)
- Amila Kahrovic
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Poschner
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Anna Schober
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Philipp Angleitner
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Leila Alajbegovic
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Martin Andreas
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Doris Hutschala
- Division of Cardiac Thoracic Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Ruud Brands
- Alloksys Life Sciences BV, 6708 PW Wageningen, The Netherlands
- IRAS Institute, University of Utrecht, 3584 Utrecht, The Netherlands
| | - Günther Laufer
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Dominik Wiedemann
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
12
|
Pickkers P, Angus DC, Arend J, Bellomo R, van den Berg E, Bernholz J, Bestle M, Broglio K, Carlsen J, Doig CJ, Ferrer R, Joannidis M, Francois B, Doi K, Kellum JA, Laterre PF, Liu K, Mehta RL, Murray PT, Ostermann M, Pettilä V, Richards S, Young P, Zarbock A, Kjølbye AL. Study protocol of a randomised, double-blind, placebo-controlled, two-arm parallel-group, multi-centre phase 3 pivotal trial to investigate the efficacy and safety of recombinant human alkaline phosphatase for treatment of patients with sepsis-associated acute kidney injury. BMJ Open 2023; 13:e065613. [PMID: 37012016 PMCID: PMC10083765 DOI: 10.1136/bmjopen-2022-065613] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2023] Open
Abstract
INTRODUCTION Sepsis, the leading cause of acute kidney injury (AKI), is associated with a high morbidity and mortality. Alkaline phosphatase (ALP) is an endogenous detoxifying enzyme. A recombinant human ALP compound, ilofotase alfa, showed no safety or tolerability concerns in a phase 2 trial. Renal function improvement over 28 days was significantly greater in the ilofotase alfa group. Moreover, a significant relative reduction in 28-day all-cause mortality of >40% was observed. A follow-up trial has been designed to confirm these findings. METHODS AND ANALYSIS This is a phase 3, global, multi-centre, randomised, double-blind, placebo-controlled, sequential design trial in which patients are randomly assigned to either placebo or 1.6 mg/kg ilofotase alfa. Randomisation is stratified by baseline modified Sequential Organ Failure Assessment (mSOFA) score and trial site. The primary objective is to confirm the survival benefit with ilofotase alfa by demonstrating a reduction in 28-day all-cause mortality in patients with sepsis-associated AKI requiring vasopressors. A maximum of 1400 patients will be enrolled at ∼120 sites in Europe, North America, Japan, Australia and New Zealand. Up to four interim analyses will take place. Based on predefined decision rules, the trial may be stopped early for futility or for effectiveness. In addition, patients with COVID-19 disease and patients with 'moderate to severe' chronic kidney disease are analysed as 2 separate cohorts of 100 patients each. An independent Data Monitoring Committee evaluates safety data at prespecified intervals throughout the trial. ETHICS AND DISSEMINATION The trial is approved by relevant institutional review boards/independent ethics committees and is conducted in accordance with the ethical principles of the Declaration of Helsinki, guidelines of Good Clinical Practice, Code of Federal Regulations and all other applicable regulations. Results of this study will determine the potential of ilofotase alfa to reduce mortality in critically ill patients with sepsis-associated AKI and will be published in a peer-reviewed scientific journal. TRIAL REGISTRATION NUMBER EudraCT CT Number 2019-0046265-24. US IND Number 117 605 Pre-results. CLINICALTRIALS gov number: NCT04411472.
Collapse
Affiliation(s)
- Peter Pickkers
- Intensive Care Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Derek C Angus
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Rinaldo Bellomo
- Department of Critical Care, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Critical Care, Austin Hospital, Melbourne, Victoria, Australia
| | | | | | - Morten Bestle
- Department of Anaesthesiology and Intensive care, Nordsjaellands Hospital, Hillerod, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Christopher J Doig
- Department of Critical Care Medicine, University of Calgary Medical Centre, Calgary, Alberta, Canada
| | - Ricard Ferrer
- Intensive Care Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Bruno Francois
- ICU and Inserm C1C, University of Limoges, Limoges, France
| | - Kent Doi
- Emergency and Critical Care Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - John A Kellum
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Kathleen Liu
- Divisions of Critical Care Medicine and Nephrology, Departments of Anesthesia and Medicine, University of California San Fransisco, San Francisco, California, USA
| | - Ravindra L Mehta
- Department of Medicine, University of California, San Diego, California, USA
| | | | - Marlies Ostermann
- Department of Critical Care, King's College London, Guy's & St Thomas' Hospital, London, UK
| | - Ville Pettilä
- Division of Intensive Care Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, Helsingin Yliopisto Laaketieteellinen tiedekunta, Helsinki, Finland
| | | | - Paul Young
- Intensive Care Unit, Wellington Hospital, Wellington, New Zealand
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, Universität Münster, Münster, Germany
| | | |
Collapse
|
13
|
Li Q, von Ehrlich-Treuenstätt V, Schardey J, Wirth U, Zimmermann P, Andrassy J, Bazhin AV, Werner J, Kühn F. Gut Barrier Dysfunction and Bacterial Lipopolysaccharides in Colorectal Cancer. J Gastrointest Surg 2023:10.1007/s11605-023-05654-4. [PMID: 36973501 PMCID: PMC10366024 DOI: 10.1007/s11605-023-05654-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/24/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND Inflammation is known to be an essential driver of various types of cancer. An increasing number of studies have suggested that the occurrence and development of colorectal cancer (CRC) are linked to the inflammatory microenvironment of the intestine. This assumption is further supported by the fact that patients with inflammatory bowel disease (IBD) are more likely to develop CRC. Multiple studies in mice and humans have shown that preoperative systemic inflammatory response is predictive of cancer recurrence after potentially curative resection. Lipopolysaccharides (LPS) are membrane surface markers of gram-negative bacteria, which induce gut barrier dysfunction and inflammation and might be significantly involved in the occurrence and development of CRC. METHODS A selective literature search was conducted in Medline and PubMed, using the terms "Colorectal Cancer", "Gut Barrier", "Lipopolysaccharides", and "Inflammation". RESULTS Disruption of intestinal homeostasis, including gut barrier dysfunction, is linked to increased LPS levels and is a critical factor for chronic inflammation. LPS can activate the diverse nuclear factor-κB (NF-κB) pathway via Toll-like receptors 4 (TLR4) to promote the inflammatory response, which aggravates gut barrier dysfunction and encourages CRC development. An intact gut barrier prevents antigens and bacteria from crossing the intestinal endothelial layer and entering circulation. In contrast, a damaged gut barrier triggers inflammatory responses and increases susceptibility to CRC. Thus, targeting LPS and the gut barrier might be a promising novel therapeutic approach for additional treatment of CRC. CONCLUSION Gut barrier dysfuction and bacterial LPS seem to play an important role in the pathogenesis and disease progression of colorectal cancer and therefore require further investigation.
Collapse
Affiliation(s)
- Qiang Li
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Viktor von Ehrlich-Treuenstätt
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Josefine Schardey
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Ulrich Wirth
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Petra Zimmermann
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Joachim Andrassy
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377, Munich, Germany
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Partner Site Munich, 81377, Munich, Germany
| | - Florian Kühn
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Marchioninistr. 15, 81377, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, 81377, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), Partner Site Munich, 81377, Munich, Germany.
| |
Collapse
|
14
|
Katasako A, Sasaki S, Raita Y, Yamamoto S, Tochitani K, Murakami M, Nishioka R, Fujisaki K. Association between serum alkaline phosphatase and bacteraemia in haemodialysis outpatients: a multicentre retrospective cross-sectional study. BMJ Open 2022; 12:e058666. [PMID: 36207044 PMCID: PMC9557305 DOI: 10.1136/bmjopen-2021-058666] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES Elevated baseline serum alkaline phosphatase (ALP) may correlate with higher medium-term to long-term mortality in the general population and in patients with chronic kidney disease. However, few data are available on the association between serum ALP and the short-term prognosis of patients on haemodialysis (HD). We verified the association of ALP levels and bacteraemia or death in maintenance HD patients suspected of bacteraemia in an outpatient setting. DESIGN We analysed 315 consecutive HD patients suspected of having bacteraemia with two sets of blood culture drawn on admission. SETTING Admission to two tertiary-care university medical centres from January 2013 to December 2015. PARTICIPANTS Consecutive cases on maintenance HD aged≥18 years. Cases of hospitalised patients who had been transferred from another hospital, had a dialysis vintage<2 months, were also undergoing peritoneal dialysis, and/or were receiving HD less than once a week were excluded. PRIMARY AND SECONDARY OUTCOME MEASURES Primary outcome measure was bacteraemia and secondary outcome was in-hospital death. RESULTS Among 315 cases included in the study, 187 had baseline-measured ALP levels, with a cut-off value on ROC analysis of 360 U/L (Area Under the Curve (AUC) 0.60, sensitivity 0.49, specificity 0.76). In multivariate analysis, there was a statistically significant association between a higher ALP in hospital visit and bacteraemia (OR: 2.37, 95% CI: 1.17 to 4.83). However, there were no statistically significant associations between higher ALP and in-hospital death (OR: 1.20, 95% CI: 0.57 to 2.54). A sensitivity analysis of 187 patients with no missing ALP values also demonstrated a significant association between elevated ALP and bacteraemia, but no significant association between ALP and in-hospital death. CONCLUSIONS Elevated ALP is a predictor of bacteraemia. In HD patients suspected of bacteraemia in outpatient settings, increased ALP levels were associated with increased likelihood of confirmed disease.
Collapse
Affiliation(s)
- Aya Katasako
- Department of Nephrology, Iizuka Hospital, Iizuka, Fukuoka, Japan
| | - Sho Sasaki
- Department of Nephrology, Iizuka Hospital, Iizuka, Fukuoka, Japan
- Clinical Research Support Office, Iizuka Hospital, Iizuka, Fukuoka, Japan
- Section of Education for Clinical Research, Kyoto University Hospital, Kyoto, Japan
- Section of Clinical Epidemiology, Department of Community Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihiko Raita
- Department of Nephrology, Okinawa Chubu Hospital, Uruma, Japan
| | - Shungo Yamamoto
- Department of Transformative Infection Control Development Studies, Osaka University Graduate School of Medicine, Osaka, Japan
- Division of Fostering Required Medical Human Resources, Center for Infectious Disease Education and Research(CiDER), Osaka University, Osaka, Japan
- Division of Infection Control and Prevention, Osaka University Hospital, Osaka, Japan
| | - Kentaro Tochitani
- Department of Healthcare Epidemiology, Kyoto University Graduate School of Public Health, Kyoto, Japan
| | - Minoru Murakami
- Department of Nephrology, Saku Central Hospital, Saku, Japan
| | - Ryo Nishioka
- Department of Rheumatology, Kanazawa University Hospital, Kanazawa, Japan
| | | |
Collapse
|
15
|
He FF, Wang YM, Chen YY, Huang W, Li ZQ, Zhang C. Sepsis-induced AKI: From pathogenesis to therapeutic approaches. Front Pharmacol 2022; 13:981578. [PMID: 36188562 PMCID: PMC9522319 DOI: 10.3389/fphar.2022.981578] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a heterogenous and highly complex clinical syndrome, which is caused by infectious or noninfectious factors. Acute kidney injury (AKI) is one of the most common and severe complication of sepsis, and it is associated with high mortality and poor outcomes. Recent evidence has identified that autophagy participates in the pathophysiology of sepsis-associated AKI. Despite the use of antibiotics, the mortality rate is still at an extremely high level in patients with sepsis. Besides traditional treatments, many natural products, including phytochemicals and their derivatives, are proved to exert protective effects through multiple mechanisms, such as regulation of autophagy, inhibition of inflammation, fibrosis, and apoptosis, etc. Accumulating evidence has also shown that many pharmacological inhibitors might have potential therapeutic effects in sepsis-induced AKI. Hence, understanding the pathophysiology of sepsis-induced AKI may help to develop novel therapeutics to attenuate the complications of sepsis and lower the mortality rate. This review updates the recent progress of underlying pathophysiological mechanisms of sepsis-associated AKI, focuses specifically on autophagy, and summarizes the potential therapeutic effects of phytochemicals and pharmacological inhibitors.
Collapse
|
16
|
Rosin DL, Hall JP, Zheng S, Huang L, Campos-Bilderback S, Sandoval R, Bree A, Beaumont K, Miller E, Larsen J, Hariri G, Kaila N, Encarnacion IM, Gale JD, van Elsas A, Molitoris BA, Okusa MD. Human Recombinant Alkaline Phosphatase (Ilofotase Alfa) Protects Against Kidney Ischemia-Reperfusion Injury in Mice and Rats Through Adenosine Receptors. Front Med (Lausanne) 2022; 9:931293. [PMID: 35966871 PMCID: PMC9366018 DOI: 10.3389/fmed.2022.931293] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/21/2022] [Indexed: 11/26/2022] Open
Abstract
Adenosine triphosphate (ATP) released from injured or dying cells is a potent pro-inflammatory "danger" signal. Alkaline phosphatase (AP), an endogenous enzyme that de-phosphorylates extracellular ATP, likely plays an anti-inflammatory role in immune responses. We hypothesized that ilofotase alfa, a human recombinant AP, protects kidneys from ischemia-reperfusion injury (IRI), a model of acute kidney injury (AKI), by metabolizing extracellular ATP to adenosine, which is known to activate adenosine receptors. Ilofotase alfa (iv) with or without ZM241,385 (sc), a selective adenosine A2A receptor (A2AR) antagonist, was administered 1 h before bilateral IRI in WT, A2AR KO (Adora2a-/- ) or CD73-/- mice. In additional studies recombinant alkaline phosphatase was given after IRI. In an AKI-on-chronic kidney disease (CKD) ischemic rat model, ilofotase alfa was given after the three instances of IRI and rats were followed for 56 days. Ilofotase alfa in a dose dependent manner decreased IRI in WT mice, an effect prevented by ZM241,385 and partially prevented in Adora2a-/- mice. Enzymatically inactive ilofotase alfa was not protective. Ilofotase alfa rescued CD73-/- mice, which lack a 5'-ectonucleotidase that dephosphorylates AMP to adenosine; ZM241,385 inhibited that protection. In both rats and mice ilofotase alfa ameliorated IRI when administered after injury, thus providing relevance for therapeutic dosing of ilofotase alfa following established AKI. In an AKI-on-CKD ischemic rat model, ilofotase alfa given after the third instance of IRI reduced injury. These results suggest that ilofotase alfa promotes production of adenosine from liberated ATP in injured kidney tissue, thereby amplifying endogenous mechanisms that can reverse tissue injury, in part through A2AR-and non-A2AR-dependent signaling pathways.
Collapse
Affiliation(s)
- Diane L. Rosin
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States,*Correspondence: Diane L. Rosin, , orcid.org/0000-0003-0187-5717
| | - J. Perry Hall
- Inflammation and Immunology Research Unit, Pfizer Inc., Cambridge, MA, United States
| | - Shuqiu Zheng
- Division of Nephrology, Center for Immunity, Inflammation and Regeneration, University of Virginia, Charlottesville, VA, United States
| | - Liping Huang
- Division of Nephrology, Center for Immunity, Inflammation and Regeneration, University of Virginia, Charlottesville, VA, United States
| | - Silvia Campos-Bilderback
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indiana Center for Biological Microscopy, Roudebush VA Medical Center, Indianapolis, IN, United States
| | - Ruben Sandoval
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indiana Center for Biological Microscopy, Roudebush VA Medical Center, Indianapolis, IN, United States
| | - Andrea Bree
- Inflammation and Immunology Research Unit, Pfizer Inc., Cambridge, MA, United States
| | - Kevin Beaumont
- BioMedicine Design, Pfizer Inc., Cambridge, MA, United States
| | - Emily Miller
- BioMedicine Design, Pfizer Inc., Groton, CT, United States
| | - Jennifer Larsen
- Early Clinical Development, Pfizer Inc., Groton, CT, United States
| | - Ghazal Hariri
- Drug Product Development, Pfizer Inc., Cambridge, MA, United States
| | - Neelu Kaila
- Medicinal Chemistry, Pfizer Inc., Cambridge, MA, United States
| | - Iain M. Encarnacion
- Division of Nephrology, Center for Immunity, Inflammation and Regeneration, University of Virginia, Charlottesville, VA, United States
| | - Jeremy D. Gale
- Inflammation and Immunology Research Unit, Pfizer Inc., Cambridge, MA, United States
| | | | - Bruce A. Molitoris
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indiana Center for Biological Microscopy, Roudebush VA Medical Center, Indianapolis, IN, United States
| | - Mark D. Okusa
- Division of Nephrology, Center for Immunity, Inflammation and Regeneration, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
17
|
Effects of Butyrate Supplementation on Inflammation and Kidney Parameters in Type 1 Diabetes: A Randomized, Double-Blind, Placebo-Controlled Trial. J Clin Med 2022; 11:jcm11133573. [PMID: 35806857 PMCID: PMC9267418 DOI: 10.3390/jcm11133573] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/27/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022] Open
Abstract
Type 1 diabetes is associated with increased intestinal inflammation and decreased abundance of butyrate-producing bacteria. We investigated the effect of butyrate on inflammation, kidney parameters, HbA1c, serum metabolites and gastrointestinal symptoms in persons with type 1 diabetes, albuminuria and intestinal inflammation. We conducted a randomized placebo-controlled, double-blind, parallel clinical study involving 53 participants randomized to 3.6 g sodium butyrate daily or placebo for 12 weeks. The primary endpoint was the change in fecal calprotectin. Additional endpoints were the change in fecal short chain fatty acids, intestinal alkaline phosphatase activity and immunoglobulins, serum lipopolysaccharide, CRP, albuminuria, kidney function, HbA1c, metabolites and gastrointestinal symptoms. The mean age was 54 ± 13 years, and the median [Q1:Q3] urinary albumin excretion was 46 [14:121] mg/g. The median fecal calprotectin in the butyrate group was 48 [26:100] μg/g at baseline, and the change was −1.0 [−20:10] μg/g; the median in the placebo group was 61 [25:139] μg/g at baseline, and the change was −12 [−95:1] μg/g. The difference between the groups was not significant (p = 0.24); neither did we find an effect of butyrate compared to placebo on the other inflammatory markers, kidney parameters, HbA1c, metabolites nor gastrointestinal symptoms. Twelve weeks of butyrate supplementation did not reduce intestinal inflammation in persons with type 1 diabetes, albuminuria and intestinal inflammation.
Collapse
|
18
|
Alkaline Phosphatase: An Old Friend as Treatment Target for Cardiovascular and Mineral Bone Disorders in Chronic Kidney Disease. Nutrients 2022; 14:nu14102124. [PMID: 35631265 PMCID: PMC9144546 DOI: 10.3390/nu14102124] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 11/17/2022] Open
Abstract
Alkaline phosphatase (ALP) is an evolutionary conserved enzyme and widely used biomarker in clinical practice. Tissue-nonspecific alkaline phosphatase (TNALP) is one of four human isozymes that are expressed as distinct TNALP isoforms after posttranslational modifications, mainly in bone, liver, and kidney tissues. Beyond the well-known effects on bone mineralization, the bone ALP (BALP) isoforms (B/I, B1, B1x, and B2) are also involved in the pathogenesis of ectopic calcification. This narrative review summarizes the recent clinical investigations and mechanisms that link ALP and BALP to inflammation, metabolic syndrome, vascular calcification, endothelial dysfunction, fibrosis, cardiovascular disease, and mortality. The association between ALP, vitamin K, bone metabolism, and fracture risk in patients with chronic kidney disease (CKD) is also discussed. Recent advances in different pharmacological strategies are highlighted, with the potential to modulate the expression of ALP directly and indirectly in CKD–mineral and bone disorder (CKD-MBD), e.g., epigenetic modulation, phosphate binders, calcimimetics, vitamin D, and other anti-fracture treatments. We conclude that the significant evidence for ALP as a pathogenic factor and risk marker in CKD-MBD supports the inclusion of concrete treatment targets for ALP in clinical guidelines. While a target value below 120 U/L is associated with improved survival, further experimental and clinical research should explore interventional strategies with optimal risk–benefit profiles. The future holds great promise for novel drug therapies modulating ALP.
Collapse
|
19
|
Wu H, Wang Y, Li H, Meng L, Zheng N, Wang J. Protective Effect of Alkaline Phosphatase Supplementation on Infant Health. Foods 2022; 11:foods11091212. [PMID: 35563935 PMCID: PMC9101100 DOI: 10.3390/foods11091212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 12/03/2022] Open
Abstract
Alkaline phosphatase (ALP) is abundant in raw milk. Because of its high heat resistance, ALP negative is used as an indicator of successful sterilization. However, pasteurized milk loses its immune protection against allergy. Clinically, ALP is also used as an indicator of organ diseases. When the activity of ALP in blood increases, it is considered that diseases occur in viscera and organs. Oral administration or injecting ALP will not cause harm to the body and has a variety of probiotic effects. For infants with low immunity, ALP intake is a good prebiotic for protecting the infant’s intestine from potential pathogenic bacteria. In addition, ALP has a variety of probiotic effects for any age group, including prevention and treatment intestinal diseases, allergies, hepatitis, acute kidney injury (AKI), diabetes, and even the prevention of aging. The prebiotic effects of alkaline phosphatase on the health of infants and consumers and the content of ALP in different mammalian raw milk are summarized. The review calls on consumers and manufacturers to pay more attention to ALP, especially for infants with incomplete immune development. ALP supplementation is conducive to the healthy growth of infants.
Collapse
Affiliation(s)
- Haoming Wu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China;
| | - Huiying Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lu Meng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Correspondence: ; Tel.: +86-10-62816069
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
20
|
Asad A, Thomas A, Dungey M, Hull KL, March DS, Burton JO. Associations between physical activity levels and renal recovery following acute kidney injury stage 3: a feasibility study. BMC Nephrol 2022; 23:140. [PMID: 35410183 PMCID: PMC9004179 DOI: 10.1186/s12882-022-02759-x] [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: 11/30/2021] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) can lead to chronic kidney disease, which results in poor long-term outcomes. There is plausibility that increased levels of physical activity may promote renal recovery post-AKI. This study aimed to investigate associations between physical activity levels and renal recovery following stage 3 AKI, and to assess the feasibility of measuring physical activity levels in this population. METHODS Forty One hospitalised patients with AKI stage 3 were enrolled. Serum creatinine and estimated glomerular filtration rate (eGFR) were collected at 12 months prior to the development of AKI, during the hospital admission when the episode of AKI stage 3 occurred, and at 1-, 3- and 6-months post discharge. All participants completed the General Practice Activity Questionnaire (GPPAQ) to assess physical activity levels. A pedometer was also worn for 7 days immediately following discharge and at 6-months post discharge to ascertain an average daily step count. Feasibility outcomes including eligibility, recruitment and retention rates, and losses to follow up were also assessed. RESULTS The average (± SD) baseline eGFR and median (IQR) serum creatinine was 71 ± 20 mL/min/1.73m2 and 85 (49) μmol/L respectively. A threefold increase in creatinine occurred during hospitalisation 436 (265) μmol/L. Greatest renal recovery occurred prior to discharge, with recovery continuing for a further three months. Inactive individuals (low GPPAQ scores) had consistently higher serum creatinine values compared to those who were active: 1 months 122 (111) μmol/L vs 70 (0) μmol/L, 6 months 112 (57) μmol/L vs 68 (0) μmol/L. Individuals with higher step counts also displayed better renal recovery 6-months post discharge (r = -0.600, p = 0.208). CONCLUSIONS Higher levels of physical activity are associated with improved renal recovery after 6- months following an episode of stage 3 AKI. A future randomised controlled trial is feasible and would be required to confirm these initial findings.
Collapse
Affiliation(s)
- Anam Asad
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Amal Thomas
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Maurice Dungey
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Katherine L Hull
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,John Walls Renal Unit, Leicester General Hospital, Leicester, UK
| | - Daniel S March
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,York Trials Unit, Department of Health Sciences, University of York, York, UK
| | - James O Burton
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK. .,John Walls Renal Unit, Leicester General Hospital, Leicester, UK. .,NIHR Leicester Biomedical Research Centre, Leicester, UK. .,School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| |
Collapse
|
21
|
Heithoff DM, Pimienta G, Mahan SP, Yang WH, Le DT, House JK, Marth JD, Smith JW, Mahan MJ. Coagulation factor protein abundance in the pre-septic state predicts coagulopathic activities that arise during late-stage murine sepsis. EBioMedicine 2022; 78:103965. [PMID: 35349828 PMCID: PMC8965145 DOI: 10.1016/j.ebiom.2022.103965] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/21/2022] [Accepted: 03/10/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Although sepsis accounts for 1 in 5 deaths globally, few molecular therapies exist for this condition. The development of effective biomarkers and treatments for sepsis requires a more complete understanding of host responses and pathogenic mechanisms at early stages of disease to minimize host-driven pathology. METHODS An alternative to the current symptom-based approach used to diagnose sepsis is a precise assessment of blood proteomic changes during the onset and progression of Salmonella Typhimurium (ST) murine sepsis. FINDINGS A distinct pattern of coagulation factor protein abundance was identified in the pre-septic state- prior to overt disease symptoms or bacteremia- that was predictive of the dysregulation of fibrinolytic and anti-coagulant activities and resultant consumptive coagulopathy during ST murine sepsis. Moreover, the changes in protein abundance observed generally have the same directionality (increased or decreased abundance) reported for human sepsis. Significant overlap of ST coagulopathic activities was observed in Gram-negative Escherichia coli- but not in Gram-positive staphylococcal or pneumococcal murine sepsis models. Treatment with matrix metalloprotease inhibitors prevented aberrant inflammatory and coagulopathic activities post-ST infection and increased survival. Antibiotic treatment regimens initiated after specific changes arise in the plasma proteome post-ST infection were predictive of an increase in disease relapse and death after cessation of antibiotic treatment. INTERPRETATION Altered blood proteomics provides a platform to develop rapid and easy-to-perform tests to predict sepsis for early intervention via biomarker incorporation into existing blood tests prompted by patient presentation with general malaise, and to stratify Gram-negative and Gram-positive infections for appropriate treatment. Antibiotics are less effective in microbial clearance when initiated after the onset of altered blood proteomics as evidenced by increased disease relapse and death after termination of antibiotic therapy. Treatment failure is potentially due to altered bacterial / host-responses and associated increased host-driven pathology, providing insight into why delays in antibiotic administration in human sepsis are associated with increased risk for death. Delayed treatment may thus require prolonged therapy for microbial clearance despite the prevailing notion of antibiotic de-escalation and shortened courses of antibiotics to improve drug stewardship. FUNDING National Institutes of Health, U.S. Army.
Collapse
Affiliation(s)
- Douglas M Heithoff
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara CA 93106, USA; Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA 93106, USA
| | - Genaro Pimienta
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Scott P Mahan
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara CA 93106, USA; Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA 93106, USA; Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis CA 95616, USA
| | - Won Ho Yang
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara CA 93106, USA; Glycosylation Network Research Center and Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; Infectious and Inflammatory Diseases Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Dzung T Le
- Department of Pathology, University of California, La Jolla, San Diego, CA 92093, USA
| | - John K House
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Camden, New South Wales 2570, Australia
| | - Jamey D Marth
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara CA 93106, USA; Infectious and Inflammatory Diseases Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Jeffrey W Smith
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Michael J Mahan
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara CA 93106, USA; Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
22
|
Fazekas B, Alagesan S, Watson L, Ng O, Conroy CM, Català C, Andres MV, Negi N, Gerlach JQ, Hynes SO, Lozano F, Elliman SJ, Griffin MD. Comparison of Single and Repeated Dosing of Anti-Inflammatory Human Umbilical Cord Mesenchymal Stromal Cells in a Mouse Model of Polymicrobial Sepsis. Stem Cell Rev Rep 2022; 18:1444-1460. [PMID: 35013938 PMCID: PMC8747454 DOI: 10.1007/s12015-021-10323-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 12/29/2022]
Abstract
Mesenchymal stromal cells (MSCs) ameliorate pre-clinical sepsis and sepsis-associated acute kidney injury (SA-AKI) but clinical trials of single-dose MSCs have not indicated robust efficacy. This study investigated immunomodulatory effects of a novel MSC product (CD362-selected human umbilical cord-derived MSCs [hUC-MSCs]) in mouse endotoxemia and polymicrobial sepsis models. Initially, mice received intra-peritoneal (i.p.) lipopolysaccharide (LPS) followed by single i.p. doses of hUC-MSCs or vehicle. Next, mice underwent cecal ligation and puncture (CLP) followed by intravenous (i.v.) doses of hUC-MSCs at 4 h or 4 and 28 h. Analyses included serum/plasma assays of biochemical indices, inflammatory mediators and the AKI biomarker NGAL; multi-color flow cytometry of peritoneal macrophages (LPS) and intra-renal immune cell subpopulations (CLP) and histology/immunohistochemistry of kidney (CLP). At 72 h post-LPS injections, hUC-MSCs reduced serum inflammatory mediators and peritoneal macrophage M1/M2 ratio. Repeated, but not single, hUC-MSC doses administered at 48 h post-CLP resulted in lower serum concentrations of inflammatory mediators, lower plasma NGAL and reversal of sepsis-associated depletion of intra-renal T cell and myeloid cell subpopulations. Hierarchical clustering analysis of all 48-h serum/plasma analytes demonstrated partial co-clustering of repeated-dose hUC-MSC CLP animals with a Sham group but did not reveal a distinct signature of response to therapy. It was concluded that repeated doses of CD362-selected hUC-MSCs are required to modulate systemic and local immune/inflammatory events in polymicrobial sepsis and SA-AKI. Inter-individual variability and lack of effect of single dose MSC administration in the CLP model are consistent with observations to date from early-phase clinical trials.
Collapse
Affiliation(s)
- Barbara Fazekas
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | | | | | - Olivia Ng
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Orbsen Therapeutics Ltd., Galway, Ireland
| | - Callum M Conroy
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Orbsen Therapeutics Ltd., Galway, Ireland
| | - Cristina Català
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | - Neema Negi
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Jared Q Gerlach
- Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Galway, Ireland
| | - Sean O Hynes
- Discipline of Pathology, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Histopathology, Galway University Hospitals, Galway, Ireland
| | - Francisco Lozano
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Servei d'Immunologia, Hospital Clínic de Barcelona, Barcelona, Spain
- Department de Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | | | - Matthew D Griffin
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland.
- Department of Nephrology, Saolta University Health Care Group, Galway University Hospitals, Galway, Ireland.
- National University of Ireland Galway, REMEDI, Biomedical Sciences, Corrib Village, Dangan, Galway, H91 TK33, Ireland.
| |
Collapse
|
23
|
Advances in pediatric acute kidney injury. Pediatr Res 2022; 91:44-55. [PMID: 33731820 DOI: 10.1038/s41390-021-01452-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/26/2021] [Accepted: 02/16/2021] [Indexed: 01/10/2023]
Abstract
The objective of this study was to inform the pediatric nephrologists of recent advances in acute kidney injury (AKI) epidemiology, pathophysiology, novel biomarkers, diagnostic tools, and management modalities. Studies were identified from PubMed, EMBASE, and Google Scholar for topics relevant to AKI. The bibliographies of relevant studies were also reviewed for potential articles. Pediatric (0-18 years) articles from 2000 to May 2020 in the English language were included. For epidemiological outcomes analysis, a meta-analysis on data regarding AKI incidence, mortality, and proportion of kidney replacement therapy was performed and an overall pooled estimate was calculated using the random-effects model. Other sections were created highlighting pathophysiology, novel biomarkers, changing definitions of AKI, evolving tools for AKI diagnosis, and various management modalities. AKI is a common condition seen in hospitalized children and the diagnosis and management have shown to be quite a challenge. However, new standardized definitions, advancements in diagnostic tools, and the development of novel management modalities have led to increased survival benefits in children with AKI. IMPACT: This review highlights the recent innovations in the field of AKI, especially in regard to epidemiology, pathophysiology, novel biomarkers, diagnostic tools, and management modalities.
Collapse
|
24
|
CSA-AKI: Incidence, Epidemiology, Clinical Outcomes, and Economic Impact. J Clin Med 2021; 10:jcm10245746. [PMID: 34945041 PMCID: PMC8706363 DOI: 10.3390/jcm10245746] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiac surgery-associated acute kidney injury (CSA-AKI) is a common complication following cardiac surgery and reflects a complex biological combination of patient pathology, perioperative stress, and medical management. Current diagnostic criteria, though increasingly standardized, are predicated on loss of renal function (as measured by functional biomarkers of the kidney). The addition of new diagnostic injury biomarkers to clinical practice has shown promise in identifying patients at risk of renal injury earlier in their course. The accurate and timely identification of a high-risk population may allow for bundled interventions to prevent the development of CSA-AKI, but further validation of these interventions is necessary. Once the diagnosis of CSA-AKI is established, evidence-based treatment is limited to supportive care. The cost of CSA-AKI is difficult to accurately estimate, given the diverse ways in which it impacts patient outcomes, from ICU length of stay to post-hospital rehabilitation to progression to CKD and ESRD. However, with the global rise in cardiac surgery volume, these costs are large and growing.
Collapse
|
25
|
Kühn F, Duan R, Ilmer M, Wirth U, Adiliaghdam F, Schiergens TS, Andrassy J, Bazhin AV, Werner J. Targeting the Intestinal Barrier to Prevent Gut-Derived Inflammation and Disease: A Role for Intestinal Alkaline Phosphatase. Visc Med 2021; 37:383-393. [PMID: 34722721 DOI: 10.1159/000515910] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/16/2021] [Indexed: 02/02/2023] Open
Abstract
Background Intestinal alkaline phosphatase (IAP) as a tissue-specific isozyme of alkaline phosphatases is predominantly produced by enterocytes in the proximal small intestine. In recent years, an increasing number of pathologies have been identified to be associated with an IAP deficiency, making it very worthwhile to review the various roles, biological functions, and potential therapeutic aspects of IAP. Summary IAP primarily originates and acts in the intestinal tract but affects other organs through specific biological axes related to its fundamental roles such as promoting gut barrier function, dephosphorylation/detoxification of lipopolysaccharides (LPS), and regulation of gut microbiota. Key Messages Numerous studies reporting on the different roles and the potential therapeutic value of IAP across species have been published during the last decade. While IAP deficiency is linked to varying degrees of physiological dysfunctions across multiple organ systems, the supplementation of IAP has been proven to be beneficial in several translational and clinical studies. The increasing evidence of the salutary functions of IAP underlines the significance of the naturally occurring brush border enzyme.
Collapse
Affiliation(s)
- Florian Kühn
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Ruifeng Duan
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Matthias Ilmer
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Ulrich Wirth
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Fatemeh Adiliaghdam
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tobias S Schiergens
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Joachim Andrassy
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Jens Werner
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| |
Collapse
|
26
|
Kalantari K, Rosner MH. Recent advances in the pharmacological management of sepsis-associated acute kidney injury. Expert Rev Clin Pharmacol 2021; 14:1401-1411. [PMID: 34493146 DOI: 10.1080/17512433.2021.1978287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Acute kidney injury is a common occurrence in patients with sepsis and portends a high mortality as well as increased morbidity with numerous sequelae including the development of chronic kidney disease. Currently, there are no specific therapies that either prevent AKI or hasten its recovery. Thus, clinicians typically rely on management of the underlying infection, optimization of hemodynamic parameters as well as avoidance of nephrotoxins to maximize outcomes. AREAS COVERED Recent advances in understanding the mechanisms of sepsis as well as how these pathways may interact to lead to acute kidney injury have opened the door to the development of new, targeted therapies. This review focuses on the operative pathways in sepsis that have been identified as critical in leading to acute kidney injury and associated therapeutic agents that target these pathways. EXPERT OPINION Despite increased understanding of the pathogenesis of sepsis, development of effective therapeutics to decrease the incidence of AKI have lagged. This is likely due to the complex pathophysiology with overlapping pathways and need for multiple therapies guided by specific biomarkers. Biomarkers that detail operative pathways may be able to guide the institution of more specific therapies with the hope for improved outcomes.
Collapse
Affiliation(s)
- Kambiz Kalantari
- Divison of Nephrology, University of Virginia Health, Charlottesville, Virginia, USA
| | - Mitchell H Rosner
- Divison of Nephrology, University of Virginia Health, Charlottesville, Virginia, USA
| |
Collapse
|
27
|
Abstract
Sepsis is a syndrome which is defined as a dysregulated host response to infection leading to organ failure. Since it remains one of the leading causes of mortality worldwide, numerous drug candidates have already been tested, and continue to be developed, as potential adjunct therapies. Despite convincing mechanisms of action and robust pre-clinical data, almost all drug candidates in the field of sepsis have failed to demonstrate clinical efficacy in the past two decades. Accordingly, the development of new sepsis drugs has markedly decreased in the past few years. Nevertheless, thanks to a better understanding of sepsis pathophysiology and pathways, new promising drug candidates are currently being developed. Instead of a unique sepsis profile as initially suspected, various phenotypes have been characterised. This has resulted in the identification of multiple targets for new drugs together with relevant biomarkers, and a better understanding of the most appropriate time to intervention. Within the entire sepsis drugs portfolio, those targeting the immune response are probably the most promising. Monoclonal antibodies targeting either cytokines or infectious agents are undoubtedly part of the potential successful therapeutic classes to come.
Collapse
Affiliation(s)
- Philippe Vignon
- Medical-Surgical Intensive Care Unit, Dupuytren Teaching Hospital, 87000, Limoges, France. .,Inserm CIC 1435, Dupuytren Teaching Hospital, 87000, Limoges, France. .,Inserm UMR 1092, Dupuytren Teaching Hospital, 87000, Limoges, France. .,Réanimation Polyvalente, CHU Dupuytren, 2 Avenue Martin Luther king, 87042, Limoges, France.
| | - Pierre-François Laterre
- St Luc University Hospital, Université Catholique de Louvain, Avenue Hippocrate 12, 1200, Brussels, Belgium
| | - Thomas Daix
- Medical-Surgical Intensive Care Unit, Dupuytren Teaching Hospital, 87000, Limoges, France.,Inserm CIC 1435, Dupuytren Teaching Hospital, 87000, Limoges, France.,Inserm UMR 1092, Dupuytren Teaching Hospital, 87000, Limoges, France
| | - Bruno François
- Medical-Surgical Intensive Care Unit, Dupuytren Teaching Hospital, 87000, Limoges, France.,Inserm CIC 1435, Dupuytren Teaching Hospital, 87000, Limoges, France.,Inserm UMR 1092, Dupuytren Teaching Hospital, 87000, Limoges, France
| |
Collapse
|
28
|
Kellum JA, Romagnani P, Ashuntantang G, Ronco C, Zarbock A, Anders HJ. Acute kidney injury. Nat Rev Dis Primers 2021; 7:52. [PMID: 34267223 DOI: 10.1038/s41572-021-00284-z] [Citation(s) in RCA: 560] [Impact Index Per Article: 186.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
Acute kidney injury (AKI) is defined by a sudden loss of excretory kidney function. AKI is part of a range of conditions summarized as acute kidney diseases and disorders (AKD), in which slow deterioration of kidney function or persistent kidney dysfunction is associated with an irreversible loss of kidney cells and nephrons, which can lead to chronic kidney disease (CKD). New biomarkers to identify injury before function loss await clinical implementation. AKI and AKD are a global concern. In low-income and middle-income countries, infections and hypovolaemic shock are the predominant causes of AKI. In high-income countries, AKI mostly occurs in elderly patients who are in hospital, and is related to sepsis, drugs or invasive procedures. Infection and trauma-related AKI and AKD are frequent in all regions. The large spectrum of AKI implies diverse pathophysiological mechanisms. AKI management in critical care settings is challenging, including appropriate volume control, nephrotoxic drug management, and the timing and type of kidney support. Fluid and electrolyte management are essential. As AKI can be lethal, kidney replacement therapy is frequently required. AKI has a poor prognosis in critically ill patients. Long-term consequences of AKI and AKD include CKD and cardiovascular morbidity. Thus, prevention and early detection of AKI are essential.
Collapse
Affiliation(s)
- John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Paola Romagnani
- Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy
| | - Gloria Ashuntantang
- Faculty of Medicine and Biomedical Sciences, Yaounde General Hospital, University of Yaounde, Yaounde, Cameroon
| | - Claudio Ronco
- Department of Medicine, University of Padova, Padua, Italy.,Department of Nephrology, Dialysis and Kidney Transplant, International Renal Research Institute, San Bortolo Hospital, Vicenza, Italy
| | - Alexander Zarbock
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
29
|
Pérez-Hernández EG, Delgado-Coello B, Luna-Reyes I, Mas-Oliva J. New insights into lipopolysaccharide inactivation mechanisms in sepsis. Biomed Pharmacother 2021; 141:111890. [PMID: 34229252 DOI: 10.1016/j.biopha.2021.111890] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/03/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
The complex pathophysiology of sepsis makes it a syndrome with limited therapeutic options and a high mortality rate. Gram-negative bacteria containing lipopolysaccharides (LPS) in their outer membrane correspond to the most common cause of sepsis. Since the gut is considered an important source of LPS, intestinal damage has been considered a cause and a consequence of sepsis. Although important in the maintenance of the intestinal epithelial cell homeostasis, the microbiota has been considered a source of LPS. Recent studies have started to shed light on how sepsis is triggered by dysbiosis, and an increased inflammatory state of the intestinal epithelial cells, expanding the understanding of the gut-liver axis in sepsis. Here, we review the gut-liver interaction in Gram-negative sepsis, exploring the mechanisms of LPS inactivation, including the recently described contribution of an isoform of the cholesteryl-ester transfer protein (CETPI). Although several key questions remain to be answered when the pathophysiology of sepsis is reviewed, new contributions coming to light exploring the way LPS might be inactivated in vivo, suggest that new applications might soon reach the clinical setting.
Collapse
Affiliation(s)
| | - Blanca Delgado-Coello
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - Ismael Luna-Reyes
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - Jaime Mas-Oliva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico.
| |
Collapse
|
30
|
Ren L, Ye J. The progress of research into the use of alkaline phosphatase in the treatment of sepsis-associated acute kidney injury. Asian J Surg 2021; 44:1212-1213. [PMID: 34210548 DOI: 10.1016/j.asjsur.2021.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022] Open
Affiliation(s)
- Liping Ren
- Dalian Medical University, Dalian, 116044, China
| | - Jilu Ye
- Department of Critical Care Medicine, Taizhou People's Hospital, Taizhou, 225300, China.
| |
Collapse
|
31
|
Larrick JW, Mendelsohn AR. Supplementation with Brush Border Enzyme Alkaline Phosphatase Slows Aging. Rejuvenation Res 2021; 23:171-175. [PMID: 32253980 DOI: 10.1089/rej.2020.2335] [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] [Indexed: 12/21/2022] Open
Abstract
Diminished integrity of the intestinal epithelial barrier with advanced age is believed to contribute to aging-associated dysfunction and pathologies in animals. In mammals, diminished gut integrity contributes to inflammaging, the increase in inflammatory processes observed in old age. Recent work suggests that expression of intestinal alkaline phosphatase (IAP) plays a key role in maintaining gut integrity. IAP expression decreases with increasing age in mice and humans. Absence of IAP leads to liver inflammation and shortened life-spans in mice lacking the IAP gene. In normal mice, exogenous supplemental IAP reverses age-induced barrier dysfunction, improves aging-associated metabolic dysfunction, prevents microbiome dysbiosis (imbalance), and extends life-span. Consistent with IAP playing a conserved role in maintaining gut integrity, increased dietary IAP increases aging-diminished physical performance in flies. IAP helps maintain gut integrity in part by supporting the expression of tight junction proteins that maintain the intestinal epithelial barrier and by inactivating bacterial pro-inflammatory factors such as lipopolysaccharides (LPS) by dephosphorylation. Recombinant IAP is in late clinical trials for sepsis-associated acute kidney injury, suggesting it may soon become available as a therapeutic. Taken together, these reports support the idea that directly increasing IAP levels by supplemental recombinant IAP or by indirectly increasing IAP levels using dietary means to induce endogenous IAP may slow the development of aging-associated pathologies.
Collapse
Affiliation(s)
- James W Larrick
- Panorama Research Institute, Sunnydale, California.,Regenerative Sciences Institute, Sunnydale, California
| | - Andrew R Mendelsohn
- Panorama Research Institute, Sunnydale, California.,Regenerative Sciences Institute, Sunnydale, California
| |
Collapse
|
32
|
Nwafor DC, Brichacek AL, Ali A, Brown CM. Tissue-Nonspecific Alkaline Phosphatase in Central Nervous System Health and Disease: A Focus on Brain Microvascular Endothelial Cells. Int J Mol Sci 2021; 22:5257. [PMID: 34067629 PMCID: PMC8156423 DOI: 10.3390/ijms22105257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/21/2022] Open
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is an ectoenzyme bound to the plasma membranes of numerous cells via a glycosylphosphatidylinositol (GPI) moiety. TNAP's function is well-recognized from earlier studies establishing its important role in bone mineralization. TNAP is also highly expressed in cerebral microvessels; however, its function in brain cerebral microvessels is poorly understood. In recent years, few studies have begun to delineate a role for TNAP in brain microvascular endothelial cells (BMECs)-a key component of cerebral microvessels. This review summarizes important information on the role of BMEC TNAP, and its implication in health and disease. Furthermore, we discuss current models and tools that may assist researchers in elucidating the function of TNAP in BMECs.
Collapse
Affiliation(s)
- Divine C. Nwafor
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA; (D.C.N.); (A.A.)
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Allison L. Brichacek
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| | - Ahsan Ali
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA; (D.C.N.); (A.A.)
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Candice M. Brown
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA; (D.C.N.); (A.A.)
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| |
Collapse
|
33
|
Praetorius H. The bacteria and the host: a story of purinergic signaling in urinary tract infections. Am J Physiol Cell Physiol 2021; 321:C134-C146. [PMID: 33979212 DOI: 10.1152/ajpcell.00054.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The local environment forces a selection of bacteria that might invade the urinary tract, allowing only the most virulent to access the kidney. Quite similar to the diet in setting the stage for the gut microbiome, renal function determines the conditions for bacteria-host interaction in the urinary tract. In the kidney, the term local environment or microenvironment is completely justified because the environment literally changes within a few micrometers. The precise composition of the urine is a function of the epithelium lining the microdomain, and the microenvironment in the kidney shows more variation in the content of nutrients, ion composition, osmolality, and pH than any other site of bacteria-host interaction. This review will cover some of the aspects of bacterial-host interaction in this unique setting and how uropathogenic bacteria can alter the condition for bacteria-host interaction. There will be a particular focus on the recent findings regarding how bacteria specifically trigger host paracrine signaling, via release of extracellular ATP and activation of P2 purinergic receptors. These finding will be discussed from the perspective of severe urinary tract infections, including pyelonephritis and urosepsis.
Collapse
|
34
|
Yang N, Wang H, Zhang R, Niu Z, Zheng S, Zhang Z. C/EBP β Mediates the Aberrant Inflammatory Response and Cell Cycle Arrest in Lps-stimulated Human Renal Tubular Epithelial Cells by Regulating NF-κB Pathway. Arch Med Res 2021; 52:603-610. [PMID: 33947580 DOI: 10.1016/j.arcmed.2021.03.008] [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: 05/16/2020] [Revised: 03/14/2021] [Accepted: 03/26/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND AND AIMS The main cause of sepsis-induced Acute kidney injury (AKI) is acute infection after surgery and subsequent progression. However, the mechanism by which AKI is caused and developed from sepsis are not completely known. Herein, we determined the role of CCAAT/enhancer-binding protein β (C/EBP β) in sepsis-induced AKI METHODS: C/EBP β expression was up or down-regulated in LPS-stimulated human renal tubular epithelial cells in vitro by recombinant adenoviruses or siRNA. Subsequent analyses included the test of TNF-α and IL-6 levels by ELISA, cell cycle assay by flow cytometry. RESULTS C/EBP β was aberrantly expressed in renal tubular epithelial HK-2 cells exposed to LPS. C/EBP β overexpression significantly enhanced, but C/EBP β silencing obviously decreased the production and secretion of inflammatory cytokines TNF-α and IL-6 induced by LPS stimulus in HK-2 cells. And the cell cycle arrest of HK-2 cells induced by LPS was also enhanced after C/EBP β overexpression while attenuated after C/EBP β silencing. Consistent pattern of changes in Cyclin D1 and p21 expression were observed in LPS-stimulated HK-2 cells after C/EBP β silencing and C/EBP β overexpression. Additionally, the increased p-NF-κB levels induced by LPS were found to be obviously decreased after C/EBP β silencing in HK-2 cells. And the enhanced TNF-α and IL-6 secretion as well as cell cycle arrest by C/EBP β overexpression were blocked by BAY11-7082 inhibitor of NF-κB pathway. CONCLUSIONS C/EBP β could mediate the LPS-induced aberrant inflammatory response and cell cycle arrest in tubular epithelial cells by NF-κB pathway.
Collapse
Affiliation(s)
- Ni Yang
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xibei Hospital, Xi'an, China
| | - Hai Wang
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xibei Hospital, Xi'an, China
| | - Rui Zhang
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xibei Hospital, Xi'an, China
| | - Zequn Niu
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xibei Hospital, Xi'an, China
| | - Shaowei Zheng
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xibei Hospital, Xi'an, China
| | - Zhengliang Zhang
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xibei Hospital, Xi'an, China.
| |
Collapse
|
35
|
Schaefer AK, Hutschala D, Andreas M, Bernardi MH, Brands R, Shabanian S, Laufer G, Wiedemann D. Decrease in serum alkaline phosphatase and prognostic relevance in adult cardiopulmonary bypass. Interact Cardiovasc Thorac Surg 2021; 31:383-390. [PMID: 32747938 DOI: 10.1093/icvts/ivaa103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/06/2020] [Accepted: 05/10/2020] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVES Cardiopulmonary bypass (CPB) induces inflammatory responses, which may lead to the loss of alkaline phosphatase (AP) that is consumed in the process of dephosphorylating detrimental extracellular nucleotides in this proinflammatory state. It has been reported that low postoperative AP levels correlate with increased postoperative support requirement and organ dysfunction after paediatric cardiac surgery. However, little is known about the perioperative development and clinical relevance of AP depletion in adults undergoing CPB. METHODS A total of 183 patients with a preoperative left ventricular ejection fraction ≤50% undergoing mitral valve surgery ± concomitant related procedures at the Department of Cardiac Surgery, Medical University of Vienna, between 2013 and 2016 were included in this retrospective analysis. Serum AP measurements at baseline and on postoperative days 1-15 were collected. Absolute and relative drop of AP on postoperative day 1 from baseline was correlated with perioperative and early postoperative parameters. Receiver operating characteristics were used to define suitable predictors and cut-offs for postoperative outcome variables. RESULTS Receiver operating characteristics showed a reduction of >50% of baseline AP to predict in-hospital mortality [area under the curve (AUC) 0.807], prolonged intensive care unit stay (>72 h, AUC 0.707), prolonged mechanical ventilation (>24 h, AUC 0.712) and surgery-related dialysis requirement (AUC 0.736). Patients with a perioperative reduction in circulating AP to levels below 50% of baseline had a significantly decreased survival. Patients with high perioperative AP loss had higher preoperative AP levels (P < 0.001), longer CPB duration (P < 0.001) and higher incidence of extracorporeal membrane oxygenation support (P < 0.001). CONCLUSIONS Increased perioperative AP loss is associated with adverse early outcome. Prospective trials are needed to determine whether this effect can be counteracted by perioperative AP supplementation.
Collapse
Affiliation(s)
- Anne-Kristin Schaefer
- Division of Cardiac Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Doris Hutschala
- Division of Cardiac Thoracic Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, Vienna, Austria
| | - Martin Andreas
- Division of Cardiac Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Martin H Bernardi
- Division of Cardiac Thoracic Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, Vienna, Austria
| | - Ruud Brands
- Alloksys Life Sciences BV, Wageningen, Netherlands
- University of Utrecht, Utrecht, Netherlands
| | - Shiva Shabanian
- Division of Cardiac Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Günther Laufer
- Division of Cardiac Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Dominik Wiedemann
- Division of Cardiac Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
36
|
Iluz-Freundlich D, Uhanova J, Grubert Van Iderstine M, Minuk GY. The impact of primary biliary cholangitis on non-alcoholic fatty liver disease. Eur J Gastroenterol Hepatol 2021; 33:565-570. [PMID: 32541239 PMCID: PMC7908858 DOI: 10.1097/meg.0000000000001782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND The impact of chronic cholestatic liver diseases such as primary biliary cholangitis (PBC) on non-alcoholic fatty liver disease (NAFLD) has yet to be described. OBJECTIVES To document and compare the severity and course of liver disease in patients with NAFLD/PBC versus NAFLD alone. METHODS In this retrospective, case-control study 68 adult NAFLD/PBC patients were matched 1:2 for age and sex with 136 NAFLD alone patients. Disease activity and severity were documented by serum aminotransferases, albumin, bilirubin and international normalized ratio (INR) values and hepatic fibrosis by Fib-4 and aspartate aminotransferase/platelet ratio indices (APRI). RESULTS On presentation (baseline), NAFLD/PBC patients had similar serum aminotransferase, albumin and bilirubin levels but lower INR values than NAFLD alone patients. Fib-4 and APRI levels were similar. Despite longer follow-up (favouring more advanced disease) in NAFLD/PBC patients, serum aminotransferases and bilirubin values were similar but albumin and INR levels significantly lower in NAFLD/PBC versus NAFLD alone patients at the end of follow-up. NAFLD/PBC patients also had significantly lower and less worsening of Fib-4 values at the end of follow-up. Transition from intermediate Fib-4 levels to those compatible with no or limited fibrosis was higher in NAFLD/PBC patients. CONCLUSION These findings suggest PBC does not adversely affect the severity or course of NAFLD.
Collapse
Affiliation(s)
- Daniel Iluz-Freundlich
- Section of Hepatology, Department of Medicine, Rady College of Medicine, University of Manitoba
| | - Julia Uhanova
- Section of Hepatology, Department of Medicine, Rady College of Medicine, University of Manitoba
| | | | - Gerald Y. Minuk
- Section of Hepatology, Department of Medicine, Rady College of Medicine, University of Manitoba
- Department of Pharmacology and Therapeutics, Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW To describe recent advances in the development of therapeutic agents for acute kidney injury (AKI). RECENT FINDINGS Traditional care for AKI is mostly supportive. At present, no specific therapy has been developed to prevent or treat AKI. However, based on a better understanding of the pathophysiology of AKI, various potential compounds have been recently identified and tested. A variety of pathways has been targeted, including oxidative and mitochondrial stress, cellular metabolism and repair, inflammation, apoptosis and hemodynamics. Many of these potential agents are currently ongoing early-phase clinical trials, and the purpose of this review is to provide a summary of those with the most potential. SUMMARY Despite the lack of therapies specifically approved for AKI, many interesting potential agents are entering clinical trials, with the potential to transform the care of patients with AKI.
Collapse
|
38
|
Juschten J, Ingelse SA, Bos LDJ, Girbes ARJ, Juffermans NP, van der Poll T, Schultz MJ, Tuinman PR. Alkaline phosphatase in pulmonary inflammation-a translational study in ventilated critically ill patients and rats. Intensive Care Med Exp 2020; 8:46. [PMID: 33336319 PMCID: PMC7746537 DOI: 10.1186/s40635-020-00335-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/31/2022] Open
Abstract
Background Alkaline phosphatase (AP), a dephosphorylating enzyme, is involved in various physiological processes and has been shown to have anti-inflammatory effects. Aim To determine the correlation between pulmonary AP activity and markers of inflammation in invasively ventilated critically ill patients with or without acute respiratory distress syndrome (ARDS), and to investigate the effect of administration of recombinant AP on pulmonary inflammation in a well-established lung injury model in rats Methods AP activity was determined and compared with levels of various inflammatory mediators in bronchoalveolar lavage fluid (BALF) samples obtained from critically ill patients within 2 days of start of invasive ventilation. The endpoints of this part of the study were the correlations between AP activity and markers of inflammation, i.e., interleukin (IL)-6 levels in BALF. In RccHan Wistar rats, lung injury was induced by intravenous administration of 10 mg/kg lipopolysaccharide, followed by ventilation with a high tidal volume for 4 h. Rats received either an intravenous bolus of 1500 IU/kg recombinant AP or normal saline 2 h after intravenous LPS administration, right before start of ventilation. Endpoints of this part of the study were pulmonary levels of markers of inflammation, including IL-6, and markers of endothelial and epithelial dysfunction. Results BALF was collected from 83 patients; 10 patients had mild ARDS, and 15 had moderate to severe ARDS. AP activity correlated well with levels of IL-6 (r = 0.70), as well as with levels of other inflammatory mediators. Pulmonary AP activity between patients with and without ARDS was comparable (0.33 [0.14–1.20] vs 0.55 [0.21–1.42] U/L; p = 0.37). Animals with acute lung injury had markedly elevated pulmonary AP activity compared to healthy controls (2.58 [2.18–3.59] vs 1.01 [0.80–1.46] U/L; p < 0.01). Intravenous administration of recombinant AP did neither affect pulmonary inflammation nor endothelial and epithelial dysfunction. Conclusions In ventilated critically ill patients, pulmonary AP activity correlates well with markers of pulmonary inflammation, such as IL-6 and IL-8. In animals with lung injury, pulmonary AP activity is elevated. Administration of recombinant AP does not alter pulmonary inflammation and endothelial or epithelial dysfunction in the acute phase of a murine lung injury model.
Collapse
Affiliation(s)
- Jenny Juschten
- Department of Intensive Care, Amsterdam University Medical Centers, location "VU", Mail stop ZH 7D-172, De Boelelaan 1117, 1082 RW, Amsterdam, the Netherlands. .,Research VUmc Intensive Care (REVIVE), Amsterdam University Medical Centers, location "VU", Amsterdam, the Netherlands. .,Department of Intensive Care, Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands. .,Laboratory of Experimental Intensive Care and Anesthesiology (L⋅E⋅I⋅C⋅A), Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands.
| | - Sarah A Ingelse
- Laboratory of Experimental Intensive Care and Anesthesiology (L⋅E⋅I⋅C⋅A), Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands.,Emma Children's Hospital-Pediatric Intensive Care Unit, Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands
| | - Lieuwe D J Bos
- Department of Pulmonology, Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands
| | - Armand R J Girbes
- Department of Intensive Care, Amsterdam University Medical Centers, location "VU", Mail stop ZH 7D-172, De Boelelaan 1117, 1082 RW, Amsterdam, the Netherlands.,Research VUmc Intensive Care (REVIVE), Amsterdam University Medical Centers, location "VU", Amsterdam, the Netherlands
| | - Nicole P Juffermans
- Laboratory of Experimental Intensive Care and Anesthesiology (L⋅E⋅I⋅C⋅A), Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands.,Department of Intensive Care, OLVG hospital, Amsterdam, The Netherlands
| | - Tom van der Poll
- Division of Infectious Diseases, Amsterdam University Medical Centers, location "AMC", Amsterdam, The Netherlands.,Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, location "AMC", Amsterdam, The Netherlands
| | - Marcus J Schultz
- Department of Intensive Care, Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands.,Laboratory of Experimental Intensive Care and Anesthesiology (L⋅E⋅I⋅C⋅A), Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands.,Mahidol-Oxford Tropical Medicine Research Unit (MORU), Mahidol University, Bangkok, Thailand.,Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Pieter Roel Tuinman
- Department of Intensive Care, Amsterdam University Medical Centers, location "VU", Mail stop ZH 7D-172, De Boelelaan 1117, 1082 RW, Amsterdam, the Netherlands.,Research VUmc Intensive Care (REVIVE), Amsterdam University Medical Centers, location "VU", Amsterdam, the Netherlands
| | | |
Collapse
|
39
|
Schippers M, Post E, Eichhorn I, Langeland J, Beljaars L, Malo MS, Hodin RA, Millán JL, Popov Y, Schuppan D, Poelstra K. Phosphate Groups in the Lipid A Moiety Determine the Effects of LPS on Hepatic Stellate Cells: A Role for LPS-Dephosphorylating Activity in Liver Fibrosis. Cells 2020; 9:E2708. [PMID: 33348845 PMCID: PMC7766276 DOI: 10.3390/cells9122708] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/21/2022] Open
Abstract
Alkaline phosphatase (AP) activity is highly upregulated in plasma during liver diseases. Previously, we demonstrated that AP is able to detoxify lipopolysaccharide (LPS) by dephosphorylating its lipid A moiety. Because a role of gut-derived LPS in liver fibrogenesis has become evident, we now examined the relevance of phosphate groups in the lipid A moiety in this process. The effects of mono-phosphoryl and di-phosphoryl lipid A (MPLA and DPLA, respectively) were studied in vitro and LPS-dephosphorylating activity was studied in normal and fibrotic mouse and human livers. The effects of intestinal AP were studied in mice with CCL4-induced liver fibrosis. DPLA strongly stimulated fibrogenic and inflammatory activities in primary rat hepatic stellate cells (rHSCs) and RAW264.7 macrophages with similar potency as full length LPS. However, MPLA did not affect any of the parameters. LPS-dephosphorylating activity was found in mouse and human livers and was strongly increased during fibrogenesis. Treatment of fibrotic mice with intravenous intestinal-AP significantly attenuated intrahepatic desmin+- and αSMA+ -HSC and CD68+- macrophage accumulation. In conclusion, the lack of biological activity of MPLA, contrasting with the profound activities of DPLA, shows the relevance of LPS-dephosphorylating activity. The upregulation of LPS-dephosphorylating activity in fibrotic livers and the protective effects of exogenous AP during fibrogenesis indicate an important physiological role of intestinal-derived AP during liver fibrosis.
Collapse
Affiliation(s)
- Marlies Schippers
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| | - Eduard Post
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| | - Ilse Eichhorn
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| | - Jitske Langeland
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| | - Leonie Beljaars
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| | - Madhu S. Malo
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (M.S.M.); (R.A.H.)
- Bangladesh Institute of Research and Rehabilitation for Diabetes, Endocrine and Metabolic Disorders (BIRDEM), Dhaka 1000, Bangladesh
| | - Richard A. Hodin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (M.S.M.); (R.A.H.)
| | - José Luis Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA;
| | - Yury Popov
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (Y.P.); (D.S.)
| | - Detlef Schuppan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (Y.P.); (D.S.)
- Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Klaas Poelstra
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| |
Collapse
|
40
|
Kellum JA, van Till JWO, Mulligan G. Targeting acute kidney injury in COVID-19. Nephrol Dial Transplant 2020; 35:1652-1662. [PMID: 33022712 PMCID: PMC7665651 DOI: 10.1093/ndt/gfaa231] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023] Open
Abstract
As of 15 August 2020, Coronavirus disease 2019 (COVID-19) has been reported in >21 million people world-wide and is responsible for more than 750,000 deaths. The occurrence of acute kidney injury (AKI) in patients hospitalized with COVID-19 has been reported to be as high as 43%. This is comparable to AKI in other forms of pneumonia requiring hospitalization, as well as in non-infectious conditions like cardiac surgery. The impact of AKI on COVID-19 outcomes is difficult to assess at present but, similar to other forms of sepsis, AKI is strongly associated with hospital mortality. Indeed, mortality is reported to be very low in COVID-19 patients without AKI. Given that AKI contributes to fluid and acid-base imbalances, compromises immune response and may impair resolution of inflammation, it seems likely that AKI contributes to mortality in these patients. The pathophysiologic mechanisms of AKI in COVID-19 are thought to be multifactorial including systemic immune and inflammatory responses induced by viral infection, systemic tissue hypoxia, reduced renal perfusion, endothelial damage and direct epithelial infection with Severe Acute Respiratory Syndrome Coronavirus 2. Mitochondria play a central role in the metabolic deregulation in the adaptive response to the systemic inflammation and are also found to be vital in response to both direct viral damage and tissue reperfusion. These stress conditions are associated with increased glycolysis and reduced fatty acid oxidation. Thus, there is a strong rationale to target AKI for therapy in COVID-19. Furthermore, many approaches that have been developed for other etiologies of AKI such as sepsis, inflammation and ischemia-reperfusion, have relevance in the treatment of COVID-19 AKI and could be rapidly pivoted to this new disease.
Collapse
Affiliation(s)
- John A Kellum
- Department of Critical Care Medicine, The Center for Critical Care Nephology, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | |
Collapse
|
41
|
Sharma A, Chakraborty R, Sharma K, Sethi SK, Raina R. Development of acute kidney injury following pediatric cardiac surgery. Kidney Res Clin Pract 2020; 39:259-268. [PMID: 32773391 PMCID: PMC7530361 DOI: 10.23876/j.krcp.20.053] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 12/30/2022] Open
Abstract
Acute kidney injury (AKI) in the pediatric population is a relatively common phenomenon. Specifically, AKI has been found in increasing numbers within the pediatric population following cardiac surgery, with up to 43% of pediatric patients developing AKI post-cardiac surgery. However, recent advances have allowed for the identification of risk factors. These can be divided into preoperative, intraoperative, and postoperative factors. Although the majority of pediatric patients developing AKI after cardiac surgery completely recover, this condition is associated with worse outcomes. These include fluid overload and increased mortality and result in longer hospital and intensive care unit stays. Detecting the presence of AKI has advanced; use of relatively novel biomarkers, including neutrophil gelatinase associated lipocalin, has shown promise in detecting more subtle changes in kidney function when compared to conventional methods. While a single, superior treatment has not been elucidated yet, novel functions of medications, including fenoldopam, theophylline and aminophylline, have been shown to have better outcomes for these patients. With the recent advances in identification of risk factors, outcomes, diagnosis, and management, the medical community can further explain the complexities of AKI in the pediatric population post-cardiac surgery.
Collapse
Affiliation(s)
- Aditya Sharma
- Department of Internal Medicine, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Ronith Chakraborty
- Department of Nephrology, Cleveland Clinic Akron General Medical Center/Akron Nephrology Associates, Akron, OH, USA
| | - Katyayini Sharma
- Department of Medicine, DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN, USA
| | - Sidharth K Sethi
- Department of Pediatric Nephrology and Pediatric Kidney Transplantation, Kidney and Urology Institute, Medanta, The Medicity Hospital, Gurgaon, India
| | - Rupesh Raina
- Department of Nephrology, Cleveland Clinic Akron General Medical Center/Akron Nephrology Associates, Akron, OH, USA.,Department of Nephrology, Akron Children's Hospital, Akron, OH, USA
| |
Collapse
|
42
|
Khailova L, Robison J, Jaggers J, Ing R, Lawson S, Treece A, Soranno D, Osorio Lujan S, Davidson JA. Tissue alkaline phosphatase activity and expression in an experimental infant swine model of cardiopulmonary bypass with deep hypothermic circulatory arrest. JOURNAL OF INFLAMMATION-LONDON 2020; 17:27. [PMID: 32817746 PMCID: PMC7422466 DOI: 10.1186/s12950-020-00256-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/30/2020] [Indexed: 01/12/2023]
Abstract
Background Infant cardiac surgery with cardiopulmonary bypass results in decreased circulating alkaline phosphatase that is associated with poor postoperative outcomes. Bovine intestinal alkaline phosphatase infusion represents a novel therapy for post-cardiac surgery organ injury. However, the effects of cardiopulmonary bypass and bovine-intestinal alkaline phosphatase infusion on tissue-level alkaline phosphatase activity/expression are unknown. Methods Infant pigs (n = 20) underwent cardiopulmonary bypass with deep hypothermic circulatory arrest followed by four hours of intensive care. Seven control animals underwent mechanical ventilation only. Cardiopulmonary bypass/deep hypothermic circulatory arrest animals were given escalating doses of bovine intestinal alkaline phosphatase infusion (0-25 U/kg/hr.; n = 5/dose). Kidney, liver, ileum, jejunum, colon, heart and lung were collected for measurement of tissue alkaline phosphatase activity and mRNA. Results Tissue alkaline phosphatase activity varied significantly across organs with the highest levels found in the kidney and small intestine. Cardiopulmonary bypass with deep hypothermic circulatory arrest resulted in decreased kidney alkaline phosphatase activity and increased lung alkaline phosphatase activity, with no significant changes in the other organs. Alkaline phosphatase mRNA expression was increased in both the lung and the ileum. The highest dose of bovine intestinal alkaline phosphatase resulted in increased kidney and liver tissue alkaline phosphatase activity. Conclusions Changes in alkaline phosphatase activity after cardiopulmonary bypass with deep hypothermic circulatory arrest and bovine intestinal alkaline phosphatase delivery are tissue specific. Kidneys, lung, and ileal alkaline phosphatase appear most affected by cardiopulmonary bypass with deep hypothermic circulatory arrest and further research is warranted to determine the mechanism and biologic importance of these changes.
Collapse
Affiliation(s)
- Ludmila Khailova
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - Justin Robison
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - James Jaggers
- Department of Surgery, University of Colorado, Aurora, CO USA
| | - Richard Ing
- Department of Anesthesiology, University of Colorado, Aurora, CO USA
| | - Scott Lawson
- Children's Hospital Colorado, Heart Institute, Aurora, CO USA
| | - Amy Treece
- Department of Pathology, University of Colorado, Aurora, CO USA
| | - Danielle Soranno
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - Suzanne Osorio Lujan
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - Jesse A Davidson
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| |
Collapse
|
43
|
Gameiro J, Fonseca JA, Outerelo C, Lopes JA. Acute Kidney Injury: From Diagnosis to Prevention and Treatment Strategies. J Clin Med 2020; 9:E1704. [PMID: 32498340 PMCID: PMC7357116 DOI: 10.3390/jcm9061704] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury (AKI) is characterized by an acute decrease in renal function that can be multifactorial in its origin and is associated with complex pathophysiological mechanisms. In the short term, AKI is associated with an increased length of hospital stay, health care costs, and in-hospital mortality, and its impact extends into the long term, with AKI being associated with increased risks of cardiovascular events, progression to chronic kidney disease (CKD), and long-term mortality. Given the impact of the prognosis of AKI, it is important to recognize at-risk patients and improve preventive, diagnostic, and therapy strategies. The authors provide a comprehensive review on available diagnostic, preventive, and treatment strategies for AKI.
Collapse
Affiliation(s)
- Joana Gameiro
- Department of Medicine, Division of Nephrology and Renal Transplantation, Centro Hospitalar Lisboa Norte, EPE, Av. Prof. Egas Moniz, 1649-035 Lisboa, Portugal
| | - José Agapito Fonseca
- Department of Medicine, Division of Nephrology and Renal Transplantation, Centro Hospitalar Lisboa Norte, EPE, Av. Prof. Egas Moniz, 1649-035 Lisboa, Portugal
| | - Cristina Outerelo
- Department of Medicine, Division of Nephrology and Renal Transplantation, Centro Hospitalar Lisboa Norte, EPE, Av. Prof. Egas Moniz, 1649-035 Lisboa, Portugal
| | - José António Lopes
- Department of Medicine, Division of Nephrology and Renal Transplantation, Centro Hospitalar Lisboa Norte, EPE, Av. Prof. Egas Moniz, 1649-035 Lisboa, Portugal
| |
Collapse
|
44
|
Acute kidney injury after nephrectomy: a new nomogram to predict postoperative renal function. BMC Nephrol 2020; 21:181. [PMID: 32410656 PMCID: PMC7227356 DOI: 10.1186/s12882-020-01839-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 05/04/2020] [Indexed: 12/15/2022] Open
Abstract
Background We aimed to develop a nomogram based on preprocedural features for early prediction of acute kidney injury (AKI) and to assess the prognosis in patients after radical and partial nephrectomy. Methods The study included a development cohort of 1111 patients who were treated between June 2012 and June 2017 and an additional validation cohort of 356 patients who were treated between July 2017 and June 2018. Stepwise regression and logistic regression analyses were used to evaluate the association between predictors and AKI. Incorporating all independent predictors, a nomogram for postoperative AKI was developed and externally validated. Patients were followed up for 5 years to assess renal function, acute kidney disease (AKD), chronic kidney disease (CKD), hospital readmission and mortality were key prognosis we focused on. Results After multivariate logistic regression, radical nephrectomy (odds ratio (OR) = 3.57, p < 0.001), aspirin (OR = 1.79, p = 0.008), systolic blood pressure (OR = 1.41, p = 0.004), triglyceride (OR = 1.26, p = 0.024), and alkaline phosphatase (OR = 1.75, p = 0.034) were independent risk factors for postoperative AKI, while albumin (OR = 0.72, p = 0.031) was a protective factor for postoperative AKI. Patients with a higher estimated glomerular filtration rate (eGFR) (60–90 ml/min/1.73 m2, OR = 0.41, p = 0.004; ≥ 90 ml/min/1.73 m2, OR = 0.37, p < 0.001) were less prone to AKI than those with a lower eGFR (< 15 ml/min/1.73 m2). These predictors were all included in the final nomogram. The area under the receiver operating characteristics curve for the model were 0.77 (p < 0.001) in the development cohort and 0.72 (p < 0.001) in the validation cohort. The incidence of AKD and CKD were 27.12 and 18.64% in AKI group, which were much higher than those in no AKI group (p < 0.001). Conclusions The nomogram had excellent predictive ability and might have significant clinical implications for the early detection of AKI in patients undergoing nephrectomy.
Collapse
|
45
|
Kühn F, Adiliaghdam F, Cavallaro PM, Hamarneh SR, Tsurumi A, Hoda RS, Munoz AR, Dhole Y, Ramirez JM, Liu E, Vasan R, Liu Y, Samarbafzadeh E, Nunez RA, Farber MZ, Chopra V, Malo MS, Rahme LG, Hodin RA. Intestinal alkaline phosphatase targets the gut barrier to prevent aging. JCI Insight 2020; 5:134049. [PMID: 32213701 PMCID: PMC7213802 DOI: 10.1172/jci.insight.134049] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/20/2020] [Indexed: 12/16/2022] Open
Abstract
Gut barrier dysfunction and gut-derived chronic inflammation play crucial roles in human aging. The gut brush border enzyme intestinal alkaline phosphatase (IAP) functions to inhibit inflammatory mediators and also appears to be an important positive regulator of gut barrier function and microbial homeostasis. We hypothesized that this enzyme could play a critical role in regulating the aging process. We tested the role of several IAP functions for prevention of age-dependent alterations in intestinal homeostasis by employing different loss-of-function and supplementation approaches. In mice, there is an age-related increase in gut permeability that is accompanied by increases in gut-derived portal venous and systemic inflammation. All these phenotypes were significantly more pronounced in IAP-deficient animals. Oral IAP supplementation significantly decreased age-related gut permeability and gut-derived systemic inflammation, resulted in less frailty, and extended lifespan. Furthermore, IAP supplementation was associated with preserving the homeostasis of gut microbiota during aging. These effects of IAP were also evident in a second model system, Drosophilae melanogaster. IAP appears to preserve intestinal homeostasis in aging by targeting crucial intestinal alterations, including gut barrier dysfunction, dysbiosis, and endotoxemia. Oral IAP supplementation may represent a novel therapy to counteract the chronic inflammatory state leading to frailty and age-related diseases in humans.
Collapse
Affiliation(s)
- Florian Kühn
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
- Department of General, Visceral and Transplant Surgery, Hospital of the University of Munich, Munich, Germany
| | - Fatemeh Adiliaghdam
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Paul M. Cavallaro
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Sulaiman R. Hamarneh
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Amy Tsurumi
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | | | - Alexander R. Munoz
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Yashoda Dhole
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Juan M. Ramirez
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Enyu Liu
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Robin Vasan
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Yang Liu
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Ehsan Samarbafzadeh
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Rocio A. Nunez
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew Z. Farber
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Vanita Chopra
- Department of Neurology,, MGH, Harvard Medical School, Boston, Massachusetts, USA
| | - Madhu S. Malo
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Laurence G. Rahme
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Richard A. Hodin
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Plaeke P, De Man JG, Smet A, Malhotra-Kumar S, Pintelon I, Timmermans JP, Nullens S, Jorens PG, Hubens G, De Winter BY. Effects of intestinal alkaline phosphatase on intestinal barrier function in a cecal ligation and puncture (CLP)-induced mouse model for sepsis. Neurogastroenterol Motil 2020; 32:e13754. [PMID: 31751495 DOI: 10.1111/nmo.13754] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Sepsis is a severe pathological condition associated with systemic inflammation, intestinal inflammation, and gastrointestinal barrier dysfunction. Intestinal alkaline phosphatase (IAP) has been demonstrated to detoxify lipopolysaccharide, an important mediator in the pathophysiology of sepsis. We investigated the effect of treatment with IAP on intestinal permeability, intestinal inflammation, and bacterial translocation. METHODS OF-1 mice were divided into 4 groups (n = 12/group), undergoing either a sham or cecal ligation and puncture (CLP) procedure to induce sepsis. Mice received IAP or a vehicle intraperitoneally 5 minutes prior to the onset of the CLP or sham procedure, which was repeated every 12 hours for two consecutive days. After two days, in vivo intestinal permeability, intestinal inflammation, and bacterial translocation were determined. KEY RESULTS CLP-induced sepsis resulted in significantly more weight loss, worse clinical disease scores, bacterial translocation, and elevated inflammatory cytokines. Intestinal permeability was increased up to 5-fold (P < .001). IAP activity was significantly increased in septic animals. Treatment with IAP had no effect on clinical outcomes but reduced the increased permeability of the small intestine by 50% (P = .005). This reduction in permeability was accompanied by a modified gene expression of claudin-1 (P = .025), claudin-14 (P = .035), and interleukin 12 (P = .015). A discriminant analysis showed that treatment with IAP is linked to modified mRNA levels of several tight junction proteins and cytokines. CONCLUSIONS AND INFERENCES Treatment with IAP diminished CLP-induced intestinal barrier disruption, associated with modified expression of several cytokines and claudins. Nevertheless, this effect did not translate into better clinical outcomes in our experimental setup.
Collapse
Affiliation(s)
- Philip Plaeke
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | - Joris G De Man
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | - Annemieke Smet
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | | | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Sara Nullens
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | - Philippe G Jorens
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium.,Department of Intensive Care Medicine, Antwerp University Hospital, Edegem (Antwerp), Belgium
| | - Guy Hubens
- Department of Abdominal Surgery, Antwerp University Hospital, Edegem (Antwerp), Belgium.,Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), University of Antwerp, Antwerp, Belgium
| | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
47
|
Singh SB, Carroll-Portillo A, Coffman C, Ritz NL, Lin HC. Intestinal Alkaline Phosphatase Exerts Anti-Inflammatory Effects Against Lipopolysaccharide by Inducing Autophagy. Sci Rep 2020; 10:3107. [PMID: 32080230 PMCID: PMC7033233 DOI: 10.1038/s41598-020-59474-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/23/2020] [Indexed: 12/21/2022] Open
Abstract
Intestinal alkaline phosphatase (IAP) regulates bicarbonate secretion, detoxifies lipopolysaccharide (LPS), regulates gut microbes, and dephosphorylates proinflammatory nucleotides. IAP also exhibits anti-inflammatory effects in a Toll-like Receptor-4 (TLR-4) dependent manner. However, it is not known whether IAP induces autophagy. We tested the hypothesis that IAP may induce autophagy which may mediate the anti-inflammatory effects of IAP. We found that exogenous IAP induced autophagy in intestinal epithelial cells and in macrophages. TLR4INC34 (C34), a TLR4 signaling inhibitor, suppressed IAP-induced autophagy. IAP also inhibited LPS-induced IL-1β mRNA expression and activation of NF-κB. When autophagy was blocked by 3-methyladenine (3MA) or by Atg5 siRNA, IAP failed to block LPS-mediated effects. IAP also upregulated autophagy-related gene expression in small intestine in mice. We administered either vehicle or IAP (100 U/ml) in drinking water for 14 days in C57BL/6 mice. Mice were sacrificed and ileal tissues collected. Increased expression of Atg5, Atg16, Irgm1, Tlr4, and Lyz genes was observed in the IAP treated group compared to the vehicle treated group. Increase in Atg16 protein expression and fluorescence intensity of LC3 was also observed in IAP-treated tissues compared to the vehicle-treated tissues. Thus, our study lays the framework for investigating how IAP and autophagy may act together to control inflammatory conditions.
Collapse
Affiliation(s)
- Sudha B Singh
- Biomedical Research Institute of New Mexico, VA Health Care System, Albuquerque, New Mexico, USA, 87108
| | - Amanda Carroll-Portillo
- Biomedical Research Institute of New Mexico, VA Health Care System, Albuquerque, New Mexico, USA, 87108
| | - Cristina Coffman
- Biomedical Research Institute of New Mexico, VA Health Care System, Albuquerque, New Mexico, USA, 87108
| | - Nathaniel L Ritz
- Biomedical Research Institute of New Mexico, VA Health Care System, Albuquerque, New Mexico, USA, 87108.,Department of Anatomy & Neuroscience, University College Cork; APC Microbiome institute, University College Cork, Cork, Ireland
| | - Henry C Lin
- Section of Gastroenterology, Medicine Service, New Mexico VA Health Care System, Albuquerque, New Mexico, USA, 87108. .,Division of Gastroenterology and Hepatology, Department of Medicine, the University of New M5052651711exico, Albuquerque, New Mexico, 87131, USA.
| |
Collapse
|
48
|
Tang W, Huang J, Huang X, Han X, Tang W, Ke G, Xu Q. Effect of alkaline phosphatase on sepsis-associated acute kidney injury patients: A systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e18788. [PMID: 31977869 PMCID: PMC7004577 DOI: 10.1097/md.0000000000018788] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND This systematic review and meta-analysis were performed to evaluate kidney function in patients with sepsis-associated acute kidney injury (SA-AKI) on alkaline phosphatase (AP) therapy. METHODS PubMed, Embase, and the Cochrane Central Register of Controlled Trials were searched electronically from inception until May 4, 2019 and randomized controlled studies assessing AP treatment in patients with SA-AKI were included. Pool analyses with fixed effects or random effects models calculated pooled mean, standard deviation, and odds ratio (OR) with 95% confidence interval (CI). RESULTS Four randomized controlled trials involving AP therapy for 392 patients with SA-AKI were included. AP had a positive effect on endogenous creatinine clearance (ECC) in patients with SA-AKI at day 14 (random effects: mean difference = 10.56, 95% CI = 2.27-18.84, P = .01) and day 28 (random effects: mean difference = 14.30, 95% CI = 6.27-22.33, P = .0005). All-cause mortality at day 28 (fixed effects: OR = 0.62, 95% CI = 0.40-0.97, P = .04) and day 90 (fixed effects: OR = 0.61, 95% CI = 0.39-0.96, P = .03) improved. Plasma creatinine level (fixed effects: mean difference = -76.83, 95% CI = -146.92 to -6.74, P = .03) and biomarkers level (random effects: mean difference = -6.57, 95% CI = -10.74 to -2.40, P < .00001) also improved in the therapy group compared with placebo. CONCLUSION In patients with SA-AKI, AP showed a relatively late protective effect by improving ECC at days 7, 14, and 28. ECC level improved when patients received AP dose of 0.212 mg/kg. Mortality improved at days 28 and 90, respectively, when patients received AP dose of 1.6 mg/kg. Levels of overall AKI biomarkers were improved in short term.
Collapse
Affiliation(s)
- Wenting Tang
- The Second Clinical College of Guangzhou University of Chinese Medicine
| | - Junlin Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangzhou
| | - Xiaowen Huang
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai
| | - Xiao Han
- First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong
| | - Wenyi Tang
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai
| | - Guibao Ke
- Department of Internal Medicine, Affiliated Hospital/Clinical Medical College of Chengdu University, Chengdu
| | - Qingqing Xu
- Department of Critical Care Medicine, Chinese Medicine Hospital of Hainan Province, Haikou, China
| |
Collapse
|
49
|
Barseghyan K, Gayer C, Azhibekov T. Differences in Serum Alkaline Phosphatase Levels in Infants with Spontaneous Intestinal Perforation versus Necrotizing Enterocolitis with Perforation. Neonatology 2020; 117:349-357. [PMID: 32750698 DOI: 10.1159/000509617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/21/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Data on laboratory markers of spontaneous intestinal perforation (SIP) and necrotizing enterocolitis (NEC) remain sparse. OBJECTIVE To compare serum alkaline phosphatase levels in infants with bowel perforation secondary to SIP versus surgical NEC, and then investigate the possible role of serum alkaline phosphatase in differentiating infants with these conditions. METHODS A retrospective case-control study of infants admitted with bowel perforation from 2005 to 2015. Demographic and prenatal data, postnatal exposures, and clinical, laboratory, and radiographic findings were extracted from inpatient medical records and analyzed using regression analysis. RESULTS Of 114 outborn infants included, 48 infants had SIP (cases) and 66 had NEC (controls). Upon admission from the referring hospital, the serum alkaline phosphatase level was significantly higher in infants with SIP, i.e., a median value of 782 versus236 U/L in NEC patients (p < 0.0001), with an adjusted odds ratio (OR) of 4.3 (p < 0.05) when the level was >500 U/L in multivariate regression model. Infants with SIP had significantly younger gestational age, presented earlier in life, primarily with pneumoperitoneum, and had greater exposure to steroids and indomethacin compared to infants with NEC. Alkaline phosphatase levels decreased rapidly in infants with SIP following admission. CONCLUSION A transient increase in serum alkaline phosphatase level is independently associated with SIP when compared to NEC. Studies to confirm the role of alkaline phosphatase in the diagnosis of SIP are necessary and have potentially significant clinical and prognostic implications.
Collapse
Affiliation(s)
- Karine Barseghyan
- Division of Neonatology, LAC+USC Medical Center/Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Kaiser Permanente Panorama City and Woodland Hills Medical Centers, Los Angeles, California, USA
| | - Christopher Gayer
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Timur Azhibekov
- Division of Neonatology, Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California Los Angeles, Los Angeles, California, USA, .,Division of Neonatology, Department of Pediatrics, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,
| |
Collapse
|
50
|
Alkaline Phosphatase Treatment of Acute Kidney Injury in an Infant Piglet Model of Cardiopulmonary Bypass with Deep Hypothermic Circulatory Arrest. Sci Rep 2019; 9:14175. [PMID: 31578351 PMCID: PMC6775126 DOI: 10.1038/s41598-019-50481-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 09/13/2019] [Indexed: 01/11/2023] Open
Abstract
Acute kidney injury (AKI) is associated with prolonged hospitalization and mortality following infant cardiac surgery, but therapeutic options are limited. Alkaline phosphatase (AP) infusion reduced AKI in phase 2 sepsis trials but has not been evaluated for cardiac surgery-induced AKI. We developed a porcine model of infant cardiopulmonary bypass (CPB) with deep hypothermic circulatory arrest (DHCA) to investigate post-CPB/DHCA AKI, measure serum/renal tissue AP activity with escalating doses of AP infusion, and provide preliminary assessment of AP infusion for prevention of AKI. Infant pigs underwent CPB with DHCA followed by survival for 4 h. Groups were treated with escalating doses of bovine intestinal AP (1, 5, or 25U/kg/hr). Anesthesia controls were mechanically ventilated for 7 h without CPB. CPB/DHCA animals demonstrated histologic and biomarker evidence of AKI as well as decreased serum and renal tissue AP compared to anesthesia controls. Only high dose AP infusion significantly increased serum or renal tissue AP activity. Preliminary efficacy evaluation demonstrated a trend towards decreased AKI in the high dose AP group. The results of this dose-finding study indicate that AP infusion at the dose of 25U/kg/hr corrects serum and tissue AP deficiency and may prevent AKI in this piglet model of infant CPB/DHCA.
Collapse
|