1
|
Lv J, Fu Z, Wang Y, Chen C, Wang Y, Zhang B, Wu H, Song Q, Li Y. Lingguizhugan decoction ameliorates renal injury secondary to heart failure by improving pyroptosis through TLR4/NF-KB/IRE1α pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156862. [PMID: 40424980 DOI: 10.1016/j.phymed.2025.156862] [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: 12/21/2024] [Revised: 04/26/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND Chronic heart failure (CHF) and chronic kidney disease (CKD) mutually promote the onset and progression of each other. Renal injury caused by heart failure currently lacks effective treatments. Previous studies have shown that Linggui Zhugan decoction (LGZGD) can significantly improve heart failure (HF) and cardiac remodeling, and has been reported to have renal protective effects. However, the effects and mechanisms of LGZGD in heart failure-induced renal injury remain unclear. PURPOSE Based on these findings, this study aims to investigate the effects and underlying mechanisms of LGZGD on renal injury secondary to HF. STUDY DESIGN We used network pharmacology to predict potential targets of LGZGD in the treatment of cardiorenal syndrome (CRS). An in vivo model of CRS with right heart failure and venous congestion was established by a single intraperitoneal injection of monocrotaline (MCT). TNF-α-stimulated NRK52E cells were used as an in vitro model. We validated the effects of LGZGD in both in vivo and in vitro experiments,. Additionally, molecular docking with the components of LGZGD identified previously was performed to predict potential targets of action. RESULTS LGZGD significantly improved heart and kidney function as well as renal histopathological changes in CRS rats. It inhibited the TLR4/NF-κB/IRE1α pathway in the kidneys and downregulated the expression of pyroptosis-related proteins (NLRP3, GSDMD, Caspase-1, IL-18, and IL-1β). Both LGZGD-containing serum and the TLR4 inhibitor (TAK-242) significantly reduced apoptosis in TNF-α-stimulated NRK52E cells and decreased the levels of TLR4/NF-κB/IRE1α pathway signaling and pyroptosis-related proteins. Molecular docking suggested that neoliquiritin (CID_51666248), enoxolon (CID_10114), and liquiritin (CID_503737) could stably bind to key targets such as IRE1α, caspase-1, and NF-κB. CONCLUSION This study demonstrated for the first time that LGZGD might ameliorate renal injury secondary to HF by improving pyroptosis through the TLR4/NF-κB/IRE1α pathway, which may provide valuable insights for future research in the treatment of CRS.
Collapse
Affiliation(s)
- Jiayu Lv
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China; Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, PR China
| | - Zhenyue Fu
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China; College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, PR China
| | - Yuxin Wang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China; Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, PR China
| | - Chunmei Chen
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China; Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, PR China
| | - Yajiao Wang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China; College of Traditional Chinese Medicine, China Academy of Chinese Medical Science, Beijing, PR China
| | - Bingxuan Zhang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Huaqin Wu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Qingqiao Song
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China.
| | - Yumeng Li
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China.
| |
Collapse
|
2
|
Ribitsch W, Lehner TA, Sauseng N, Rosenkranz AR, Schneditz D. Susceptibility of hepato-splanchnic perfusion to intra-abdominal pressure in peritoneal dialysis patients. Perit Dial Int 2024:8968608241275922. [PMID: 39196595 DOI: 10.1177/08968608241275922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND The impact of peritoneal filling on hepato-splanchnic perfusion during peritoneal dialysis has not been fully elucidated yet. METHODS Measurements were done in 20 prevalent peritoneal dialysis patients during a peritoneal equilibration test (PET) with 2L of standard dialysate. Data were obtained in the drained state at baseline (T0), after instillation (T1), and after 2 h of dwell time (T2). Intra-abdominal pressure (IAP) was measured by Durand's approach. The hepatic clearance index (KI) of indocyanine-green (ICG) was determined as an indirect measure of hepato-splanchnic blood flow. Cardiac index (CI), heart rate (HR), and total peripheral resistance index (TPRI) were derived from continuous arterial pulse analysis. Fluid volume overload (VO) was evaluated by multifrequency bioimpedance analysis. Ejection fraction (EF) was obtained from echocardiographic examination. RESULTS IAP was 5.8 ± 3.5 mmHg at baseline (T0), rose to 9.4 ± 2.8 mmHg after instillation of dialysate (T1), and further to 9.7 ± 2.8 mmHg after 2 h of dwell time (p < 0.001). KI slightly declined from 0.60 ± 0.22 L/min/m2 at T0 to 0.53 ± 0.15 L/min/m2 at T1 (p = 0.075), and returned to 0.59 ± 0.22 L/min/m2 at T2 (p = 0.052). CI, HR, and TPRI did not change significantly. In five patients with an EF < 40% KI was significantly lower at T1 (0.42 ± 0.12 L/min/m2; p = 0.039), and further decreased at T2 (0.40 ± 0.04 L/min/m2; p = 0.016) compared to patients with normal EF (T1: 0.58 ± 0.15 L/min/m2 and T2: 0.67 ± 0.22 L/min/m2). CONCLUSIONS Overall, hepatic clearance of ICG as a marker of hepato-splanchnic blood flow is not affected by the filling of the peritoneal cavity.
Collapse
Affiliation(s)
- Werner Ribitsch
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Thomas A Lehner
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria
| | - Notburga Sauseng
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria
| | - Alexander R Rosenkranz
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Daniel Schneditz
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria
| |
Collapse
|
3
|
Zhang S, Li S, Gao L, Zhao Q, Yang T, Zeng Q, Huang Z, Li X, Duan A, Wang Y, Zhao Z, Luo Q, Liu Z. Effects of malnutrition on disease severity and adverse outcomes in idiopathic pulmonary arterial hypertension: a retrospective cohort study. Respir Res 2024; 25:292. [PMID: 39080722 PMCID: PMC11290113 DOI: 10.1186/s12931-024-02925-9] [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: 05/22/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Malnutrition is common in patients with chronic cardiovascular disease and is associated with significantly higher all-cause mortality. Approximately one-third of patients with heart failure are malnourished. However, the relationship between malnutrition and idiopathic pulmonary arterial hypertension (IPAH) remains unclear. This study aimed to clarify the prognostic value of malnutrition in patients with IPAH. METHODS A total of 432 consecutive participants with IPAH were included in this study between March 2013 and August 2021. Three common malnutrition assessment tools, including the geriatric nutritional risk index (GNRI), prognostic nutritional index (PNI), and controlling nutritional status (CONUT) score, were used to evaluate the nutritional status of patients with IPAH. The relationships between the malnutrition tools and long-term adverse outcomes were determined using restricted cubic splines and multivariate Cox regression models. RESULTS During a mean follow-up of 3.1 years, 158 participants experienced clinical worsening or all-cause death. Patients were stratified into the low-, intermediate- and high-risk groups based on the European Society of Cardiology (ESC) risk stratification, and the PNI (55.9 ± 5.7 vs. 54.4 ± 7.2 vs. 51.1 ± 7.1, P = 0.005) and CONUT score (2.1 ± 0.9 vs. 2.5 ± 1.2 vs. 3.3 ± 1.1, P < 0.001) identified these patient groups better than the GNRI. All three malnutrition tools were associated with well-validated variables that reflected IPAH severity, such as the World Health Organization functional class, 6-min walk distance, and N-terminal pro-brain natriuretic peptide level. The CONUT score exhibited better predictive ability than both the GNRI (ΔAUC = 0.059, P < 0.001) and PNI (ΔAUC = 0.095, P < 0.001) for adverse outcomes and significantly improved reclassification and discrimination beyond the ESC risk score. Multivariable Cox regression analysis indicated that only the CONUT score (hazard ratio = 1.363, 95% confidence interval 1.147, 1.619 per 1.0-standard deviation increment, P < 0.001) independently predicted adverse outcomes. CONCLUSIONS The malnutrition status was associated with disease severity in patients with IPAH. The CONUT score provided additional information regarding the risk of clinically worsening events, making it a meaningful risk stratification tool for these patients.
Collapse
Affiliation(s)
- Sicheng Zhang
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Sicong Li
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Luyang Gao
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Qing Zhao
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Tao Yang
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Qixian Zeng
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Zhihua Huang
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Xin Li
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Anqi Duan
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yijia Wang
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Zhihui Zhao
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China.
| | - Qin Luo
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China.
| | - Zhihong Liu
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China.
| |
Collapse
|
4
|
McIntyre CW. Update on Hemodialysis-Induced Multiorgan Ischemia: Brains and Beyond. J Am Soc Nephrol 2024; 35:653-664. [PMID: 38273436 PMCID: PMC11149050 DOI: 10.1681/asn.0000000000000299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/17/2023] [Indexed: 01/27/2024] Open
Abstract
Hemodialysis is a life-saving treatment for patients with kidney failure. However, patients requiring hemodialysis have a 10-20 times higher risk of cardiovascular morbidity and mortality than that of the general population. Patients encounter complications such as episodic intradialytic hypotension, abnormal perfusion to critical organs (heart, brain, liver, and kidney), and damage to vulnerable vascular beds. Recurrent conventional hemodialysis exposes patients to multiple episodes of circulatory stress, exacerbating and being aggravated by microvascular endothelial dysfunction. This promulgates progressive injury that leads to irreversible multiorgan injury and the well-documented higher incidence of cardiovascular disease and premature death. This review aims to examine the underlying pathophysiology of hemodialysis-related vascular injury and consider a range of therapeutic approaches to improving outcomes set within this evolved rubric..
Collapse
Affiliation(s)
- Christopher W McIntyre
- Lilibeth Caberto Kidney Clinical Research Unit, Lawson Health Research Institute, London, Ontario, Canada, and Departments of Medicine, Medical Biophysics and Pediatrics, Western University, London, Ontario, Canada
| |
Collapse
|
5
|
Foster DM, Kellum JA. Endotoxic Septic Shock: Diagnosis and Treatment. Int J Mol Sci 2023; 24:16185. [PMID: 38003374 PMCID: PMC10671446 DOI: 10.3390/ijms242216185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Endotoxin, also referred to as lipopolysaccharide (LPS), is a potent stimulator of the inflammatory cascade which may progress to sepsis and septic shock. The term endotoxic septic shock has been used for patients who have a clinical phenotype that is characterized by high endotoxin activity in addition to a high burden of organ failure; especially a pattern of organ failure including hepatic dysfunction, acute kidney injury, and various forms of endothelial dysfunction. Endotoxic septic shock has been a target for drug therapy for decades with no success. A likely barrier to their success was the inability to quantify endotoxin in the bloodstream. The Endotoxin Activity Assay (EAA) is positioned to change this landscape. In addition, medical devices using adsorptive technology in an extra-corporeal circulation has been shown to remove large quantities of endotoxin from the bloodstream. Focusing on the use of EAA to determine high concentrations of endotoxin will allow patients with endotoxic septic shock to be identified quickly and these patients may benefit most from removal of endotoxin using extracorporeal methods.
Collapse
Affiliation(s)
| | - John A. Kellum
- Spectral Medical Inc., Toronto, ON M9C 1C2, Canada;
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
6
|
Xie Z, Yang C, Xu T. Hesperetin attenuates LPS-induced the inflammatory response and apoptosis of H9c2 by activating the AMPK/P53 signaling pathway. Immun Inflamm Dis 2023; 11:e973. [PMID: 37584301 PMCID: PMC10413818 DOI: 10.1002/iid3.973] [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: 02/12/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/17/2023] Open
Abstract
INTRODUCTION Hesperetin (HES), whose main pharmacological effects are anti-inflammatory and cardioprotective properties. In our study, we investigated the role of HES in lipopolysaccharide (LPS)-induced inflammation and apoptosis in H9c2 cells. METHODS Cell viability was assessed through MTT assay. Tumor necrosis factor (TNF)-α and interleukin (IL)-β expression were quantified through RT-qPCR assay. Secondly, the apoptosis rate was assessed by Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay. Finally, B-cell lymphoma 2 (Bcl-2)- associated X protein (Bax), adenosine monophosphate-activated protein kinase (AMPK), and P53 expression were quantified through western blot assay. RESULTS Our results demonstrated that LPS stimulation decreased the cell viability, increased IL-1β and TNF-α expression in H9c2 cells. However, HES treatment significantly increased the cell viability, decreased IL-1β and TNF-α expression in LPS-induced H9c2 cells. In addition, HES significantly increased the phosphorylation level of AMPK. Meanwhile, HES prevented against LPS-mediated the P53 and Bax protein upregulation, and Bcl-2 protein downregulation in H9c2 cells. More interestingly, compound C (an AMPK inhibitor) treatment eliminated the protective effects of HES. CONCLUSION Our findings revealed that HES attenuated the LPS-mediated inflammation and apoptosis of H9c2 cells by activating the AMPK/P53 signaling pathway, suggesting that HES may be a potential cardioprotective agent.
Collapse
Affiliation(s)
- Zan Xie
- Department of Cardiologythe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiShandongChina
| | - Chunxia Yang
- Department of Cardiologythe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiShandongChina
| | - Tingting Xu
- Department of Cardiologythe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiShandongChina
| |
Collapse
|
7
|
Olek K, Kuczaj AA, Warwas S, Hrapkowicz T, Przybyłowski P, Tanasiewicz M. Gut Microbiome in Patients after Heart Transplantation-Current State of Knowledge. Biomedicines 2023; 11:1588. [PMID: 37371683 DOI: 10.3390/biomedicines11061588] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/13/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
The human gut microbiota include over 10 trillion microorganisms, such as bacteria, fungi, viruses, archaea, and protozoa. Many reports indicate the strong correlation between dysbiosis and the severity of cardiovascular diseases. Microbiota seem to interact with the host's alloimmunity and may have an immunomodulatory role in graft rejection processes. In our study, we present the current state of the knowledge of microbiota in heart transplant recipients. We present up-to-date microbiota diagnostic methods, interactions between microbiota and immunosuppressive drugs, the immunomodulatory effects of dysbiosis, and the available strategies (experimental and clinical strategies) to modulate host microbiota.
Collapse
Affiliation(s)
- Katarzyna Olek
- Department of Dental Propedeutics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Zabrze, Poland
| | - Agnieszka Anna Kuczaj
- Department of Cardiac Surgery, Transplantology, Vascular and Endovascular Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, M.C. Sklodowskiej 9, 41-800 Zabrze, Poland
| | - Szymon Warwas
- Students' Scientific Association Affiliated with the Department of Cardiac, Vascular and Endovascular Surgery and Transplantology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Zabrze, Poland
| | - Tomasz Hrapkowicz
- Department of Cardiac Surgery, Transplantology, Vascular and Endovascular Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, M.C. Sklodowskiej 9, 41-800 Zabrze, Poland
| | - Piotr Przybyłowski
- Department of Cardiac Surgery, Transplantology, Vascular and Endovascular Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, M.C. Sklodowskiej 9, 41-800 Zabrze, Poland
| | - Marta Tanasiewicz
- Department of Conservative Dentistry and Endodontics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Zabrze, Poland
| |
Collapse
|
8
|
Matsiras D, Bezati S, Ventoulis I, Verras C, Parissis J, Polyzogopoulou E. Gut Failure: A Review of the Pathophysiology and Therapeutic Potentials in the Gut-Heart Axis. J Clin Med 2023; 12:2567. [PMID: 37048650 PMCID: PMC10095379 DOI: 10.3390/jcm12072567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Despite considerable advances in the field, heart failure (HF) still poses a significant disease burden among affected individuals since it continues to cause high morbidity and mortality rates. Inflammation is considered to play a key role in disease progression, but the exact underlying pathophysiological mechanisms involved have not yet been fully elucidated. The gut, as a potential source of inflammation, could feasibly explain the state of low-grade inflammation seen in patients with chronic HF. Several derangements in the composition of the microbiota population, coupled with an imbalance between favorable and harmful metabolites and followed by gut barrier disruption and eventually bacterial translocation, could contribute to cardiac dysfunction and aggravate HF. On the other hand, HF-associated congestion and hypoperfusion alters intestinal function, thereby creating a vicious cycle. Based on this evidence, novel pharmaceutical agents have been developed and their potential therapeutic use has been tested in both animal and human subjects. The ultimate goal in these efforts is to reverse the aforementioned intestinal derangements and block the inflammation cascade. This review summarizes the gut-related causative pathways implicated in HF pathophysiology, as well as the associated therapeutic interventions described in the literature.
Collapse
Affiliation(s)
- Dionysis Matsiras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Sofia Bezati
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, 50200 Ptolemaida, Greece
| | - Christos Verras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - John Parissis
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Effie Polyzogopoulou
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| |
Collapse
|
9
|
Yuzefpolskaya M, Bohn B, Ladanyi A, Khoruts A, Colombo PC, Demmer RT. Oral and gut microbiome alterations in heart failure: Epidemiology, pathogenesis and response to advanced heart failure therapies. J Heart Lung Transplant 2023; 42:291-300. [PMID: 36586790 DOI: 10.1016/j.healun.2022.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/18/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Despite significant advances in therapies, heart failure (HF) remains a progressive disease that, once advanced, is associated with significant death and disability. Cardiac replacement therapies with left ventricular assist device (LVAD) and heart transplantation (HT) are the only treatment options for advanced HF, while lifesaving they can also be lifespan limiting due to the associated complications. Systemic inflammation is mechanistically important in HF pathophysiology and progression. However, directly targeting inflammation in HF has not been beneficial thus far. These failed attempts at therapeutics might be related to our limited understanding of the factors that cause inflammation in HF, and, therefore, to our inability to investigate these triggers in interventional studies. Observational studies have consistently demonstrated associations between alterations in the digestive (gut and oral) microbiome, inflammation and HF risk and progression. Additionally, recent data indicate that these microbial perturbations persist following LVAD and HT, along with residual inflammation and oxidative stress. Furthermore, there is rising recognition of the critical contribution of the microbiome to the metabolism of immunosuppressive drugs after HT. Cumulatively, these findings might posit a mechanistic link between microbiome alterations, systemic inflammation, and adverse outcomes in HF patients before and after cardiac replacement therapies. This review (1) provides an update on available data linking changes in digestive tract microbiota, inflammation, and oxidative stress, to HF pathogenesis and progression; (2) describes evolution of these relationships following LVAD and HT; and (3) outlines present and future intervention strategies that can manipulate the microbiome and possibly modify HF disease trajectory.
Collapse
Affiliation(s)
- Melana Yuzefpolskaya
- Division of Cardiovascular Medicine, Columbia University Irving Medical Center, New York City, New York.
| | - Bruno Bohn
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Annamaria Ladanyi
- Division of Cardiovascular Medicine, Columbia University Irving Medical Center, New York City, New York
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine University of Minnesota, Minneapolis, Minnesota
| | - Paolo C Colombo
- Division of Cardiovascular Medicine, Columbia University Irving Medical Center, New York City, New York
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota; Division of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
10
|
Loosen SH, Krieg S, Gaensbacher J, Doege C, Krieg A, Luedde T, Luedde M, Roderburg C, Kostev K. The Association between Antibiotic Use and the Incidence of Heart Failure: A Retrospective Case-Control Study of 162,188 Outpatients. Biomedicines 2023; 11:biomedicines11020260. [PMID: 36830796 PMCID: PMC9953253 DOI: 10.3390/biomedicines11020260] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
The pathogenesis of heart failure (HF) is multifactorial, and is characterized by structural, cellular, and molecular remodeling processes. Inflammatory signaling pathways may play a particularly understudied role in HF. Recent data suggest a possible impact of antibiotic use on HF risk. Therefore, the aim of this retrospective case-control study was to investigate the association between antibiotic use and the incidence of HF. Data from the Disease Analyzer (IQVIA) database for patients diagnosed with HF and matched non-HF controls from 983 general practices in Germany between 2000 and 2019 were analyzed. A multivariable conditional logistic regression model was performed. Regression models were calculated for all patients, as well as for data stratified for sex and four age groups. A total of 81,094 patients with HF and 81,094 patients without HF were included in the analyses. In the regression analysis, low, but not high, total antibiotic use was significantly associated with a slightly lower HF risk compared with non-antibiotic users (OR: 0.87; 95% CI: 0.85-0.90). A significantly lower HF incidence was observed for sulfonamides and trimethoprim (OR: 0.87, 95% CI: 0.81-0.93) and for macrolides (OR: 0.87, 95% CI: 0.84-0.91). High use of cephalosporins, however, was associated with an increased HF risk (OR: 1.16; 95% CI: 1.11-1.22). In conclusion, this study from a large real-world cohort from Germany provides evidence that the use of different antibiotics may be associated with HF risk in a dose-dependent manner, possibly due to involved inflammatory processes. Overall, this study should provide a basis for future research to offer new therapeutic strategies for HF patients to improve their limited prognosis.
Collapse
Affiliation(s)
- Sven H. Loosen
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty of Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Sarah Krieg
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty of Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Julia Gaensbacher
- Internal Medicine III, University of Schleswig Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Corinna Doege
- Department of Pediatric Neurology, Center of Pediatrics and Adolescent Medicine, Central Hospital Bremen, 28211 Bremen, Germany
| | - Andreas Krieg
- Department of Surgery (A), University Hospital Duesseldorf, Medical Faculty of Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty of Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Mark Luedde
- Internal Medicine III, University of Schleswig Holstein, Campus Kiel, 24105 Kiel, Germany
- KGP Bremerhaven, 27574 Bremerhaven, Germany
- Correspondence: (M.L.); (C.R.); Tel.: +49-471-309-6090 (M.L.); Fax: +49-471-309-6099 (M.L.)
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty of Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
- Correspondence: (M.L.); (C.R.); Tel.: +49-471-309-6090 (M.L.); Fax: +49-471-309-6099 (M.L.)
| | | |
Collapse
|
11
|
Muacevic A, Adler JR. Risk Factors and Outcomes of Protein-Calorie Malnutrition in Chronic Heart Failure Patients Undergoing Elective Cardiac Surgery. Cureus 2022; 14:e30378. [PMID: 36407213 PMCID: PMC9667748 DOI: 10.7759/cureus.30378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION Chronic heart failure (CHF) patients are often malnourished. Our aim was to determine the effect of protein-calorie malnutrition (PCM) on in-hospital outcomes in CHF patients following elective cardiac surgery and to identify risk factors for PCM in this patient population. METHODS A retrospective analysis of the National Inpatient Sample (NIS) database was conducted from 2016 to 2018. In-hospital outcomes in adult patients with CHF undergoing elective coronary artery bypass graft (CABG) with cardiopulmonary bypass-assist or cardiac valve replacement surgeries were analyzed. Propensity-score matching was used to match CHF patients with and without PCM and followed by logistic regression analysis. A multivariate logistic regression model was used to identify the risk factors associated with PCM in this population. RESULTS In total 25,940 CHF patients were identified, of which 6,271 underwent elective CABG and 19,669 underwent valve replacement surgeries. The prevalence of PCM in CHF patients undergoing CABG and valve replacement was 3.9% and 2.9%, respectively. CHF patients with PCM had significantly higher risk of in-hospital mortality, post-operative cardiac and gastrointestinal complications compared to CHF patients without PCM. The mean hospital length of stay was twice as high in the PCM group (mean days: 18.6 vs 9.9). Female gender, Black race (vs White race), a high Charlson Comorbidity Index, Medicare/Medicaid insurance status (vs private insurance), and CHF (systolic and combined systolic and diastolic) were independently associated with significantly higher risk of PCM diagnosis. CONCLUSIONS CHF patients with PCM who undergo elective CABG or valve replacement surgeries are at a significantly higher risk of mortality, post-operative cardiac and gastrointestinal complications, and increased duration of hospital stay compared to those without PCM. Future prospective studies should assess the CHF patients who are at a higher risk of PCM and whether correcting pre-operative nutrition in this surgical population can improve outcomes following cardiac surgery.
Collapse
|
12
|
The Effects of Bacterial Lipopolysaccharide (LPS) on Turkey Poults: Assessment of Biochemical Parameters and Histopathological Changes. Vet Sci 2022; 9:vetsci9050240. [PMID: 35622768 PMCID: PMC9146353 DOI: 10.3390/vetsci9050240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 01/30/2023] Open
Abstract
A lipopolysaccharide (LPS) is a large molecule and an outer membrane glycolipid found in Gram-negative bacteria, including Escherichia coli (E. coli). These molecules (LPS) target acute inflammatory responses and significant physiological changes. Importantly, E. coli is considered one of the most important bacterial causes of avian colibacillosis that affect domestic turkey industry. However, little information is available about the potential influence of LPS on the biochemical parameters and histopathological changes in turkey poults. Therefore, this study aimed to evaluate the influence of bacterial lipopolysaccharide (LPS) molecules on serum biomarkers and histopathological changes in turkey poults. The birds were randomly divided into five groups, as follows: group I did not receive any inoculation; group II was inoculated with sterile saline; and groups III, IV, and V were inoculated intraperitoneally with LPS at 0.01, 0.1, and 1 mg/kg of body weight (BW), respectively. The biochemical parameters and the histopathology of different organs were examined in all birds one day post-inoculation. Our results revealed hypolipidemia, hypoglycemia, a significant decrease in uric acid, and a significant increase in serum activities of aspartate transaminase (AST), alanine transaminase (ALT), alkaline phosphatase (ALP), lactate dehydrogenase (LDH), and creatine kinase (CK), as well as cardiac troponin T concentrations in treated groups. Moreover, there was a significant increase in α1-, β-, and γ-globulin concentrations and a decrease in albumin and α2-globulin concentrations in group V. However, a significant increase in α2- and γ-globulin levels and a decrease in albumin levels were detected in groups III and IV. In addition, significant decreases in the albumin/globulin ratio were recorded in all LPS-treated groups. Hepatocellular and cardiac muscle necrosis, slight renal changes, and massive pulmonary inflammatory reactions were recorded. This study provides valuable information about serum biomarkers, protein fractions, and histopathological changes in turkey poults treated with LPS for further investigations of pathophysiological mechanisms in avian medicine along with biomedical research.
Collapse
|
13
|
Jarry S, Calderone A, Dion D, Bouchard D, Couture ÉJ, Denault A. Acute Cardiointestinal Syndrome Resulting From Postoperative Acute Biventricular Heart Failure. J Cardiothorac Vasc Anesth 2022; 36:2220-2227. [PMID: 35331631 DOI: 10.1053/j.jvca.2022.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/08/2022] [Indexed: 01/16/2023]
Affiliation(s)
- Stéphanie Jarry
- Department of Anesthesiology, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Alexander Calderone
- Department of Anesthesiology, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Daniel Dion
- Department of Pathology and Cell Biology, Hôpital du Sacré-Coeur de Montréal, Montreal, Canada
| | - Denis Bouchard
- Department of Cardiac Surgery, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Étienne J Couture
- Department of Anesthesiology and Department of Medicine, Division of Intensive Care Medicine, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada
| | - André Denault
- Department of Anesthesiology, Montreal Heart Institute, Université de Montréal, Montreal, Canada; Division of Critical Care, Montreal Heart Institute, Université de Montréal, Montreal, Canada.
| |
Collapse
|
14
|
Lv S, Wang Y, Zhang W, Shang H. Trimethylamine oxide: a potential target for heart failure therapy. Heart 2021; 108:917-922. [PMID: 34611047 DOI: 10.1136/heartjnl-2021-320054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/09/2021] [Indexed: 11/04/2022] Open
Abstract
Heart failure (HF) is a clinical syndrome in the late stage of cardiovascular disease and is associated with high prevalence, mortality and rehospitalisation rate. The pathophysiological mechanisms of HF have experienced the initial 'water-sodium retention' mode to 'abnormal hemodynamics' mode, and subsequent to 'abnormal activation of neuroendocrine' mode, which has extensively promoted the reform of HF treatment and updated the treatment concept. Since the Human Microbiome Project commencement, the study on intestinal microecology has swiftly developed, providing a new direction to reveal the occurrence of diseases and the mechanisms behind drug effects. Intestinal microecology comprises the gastrointestinal lumen, epithelial secretion, food entering the intestine, intestinal flora and metabolites. Choline and L-carnitine in the diet are metabolised to trimethylamine (TMA) by the intestinal micro-organisms, with TMA being absorbed into the blood. TMA then enters the liver through the portal vein circulation and is oxidised to trimethylamine oxide (TMAO) by the hepatic flavin-containing mono-oxygenase (FMO) family, especially FMO3. The circulating TMAO levels are associated with adverse outcomes in HF (mortality and readmission), and lower TMAO levels indicate better prognosis. As HF progresses, the concentration of TMAO in patients gradually increases. Whether the circulating TMAO level can be decreased by intervening with the intestinal microflora or relevant enzymes, thereby affecting the prognosis of patients with HF, has become a research hotspot. Therefore, based on the HF intestinal hypothesis, exploring the treatment strategy for HF targeting the TMAO metabolite of the intestinal flora may update the treatment concept in HF and improve its therapeutic effect.
Collapse
Affiliation(s)
- Shichao Lv
- Key Laboratory of Chinese Internal Medicine of MOE, Dongzhimen Hospital, BUCM, Beijing, China.,First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunjiao Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wanqin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of MOE, Dongzhimen Hospital, BUCM, Beijing, China
| |
Collapse
|
15
|
Yang L, Chen C, Lv B, Gao Y, Li G. Epoxyeicosatrienoic acids prevent cardiomyocytes against sepsis by A 2AR-induced activation of PI3K and PPARγ. Prostaglandins Other Lipid Mediat 2021; 157:106595. [PMID: 34597782 DOI: 10.1016/j.prostaglandins.2021.106595] [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: 03/30/2021] [Revised: 08/14/2021] [Accepted: 09/24/2021] [Indexed: 10/20/2022]
Abstract
Although epoxyeicosatrienoic acids (EETs) have multiple protective effects against different diseases, whether they can improve the pathogenesis of lipopolysaccharide (LPS)-induced septic cardiac dysfunction remains unknown. We investigated the effects of EETs on the LPS-induced inflammatory response in myocardial dysfunction mice and H9c2 cardiac myocytes. Cardiac-specific CYP2J2 transgenic mice (Tr) showed improved cardiac function and reduced inflammation response after administration with LPS, while the protective effects were not observed in A2A adenosine receptor (A2AR/ADORA2A)-deficient mice (knockout/KO). In vitro, EETs prevented LPS-induced inflammation and apoptosis in the cardiomyocytes via A2AR activation. Moreover, ZM241385 (A2AR inhibitor) attenuated the cardioprotective properties of EETs. Further investigation demonstrated that A2AR signal pathway activation partly regulated phosphatidylinositol 3-kinase (PI3K) and peroxisome proliferator-activated receptor-γ (PPARγ) expression. This is the first report on EETs exerting cardioprotective effects against LPS-induced cardiomyocyte injury via A2AR activation.
Collapse
Affiliation(s)
- Lei Yang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Chen Chen
- Departments of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Bingya Lv
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Yi Gao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China.
| |
Collapse
|
16
|
Mitchell JA, Kirkby NS, Ahmetaj-Shala B, Armstrong PC, Crescente M, Ferreira P, Lopes Pires ME, Vaja R, Warner TD. Cyclooxygenases and the cardiovascular system. Pharmacol Ther 2021; 217:107624. [DOI: 10.1016/j.pharmthera.2020.107624] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023]
|
17
|
Abstract
Cardiorenal syndrome is a complex interplay of dysregulated heart and kidney interaction that leads to multiorgan system dysfunction, which is not an uncommon occurrence in the setting of right heart failure. The traditional concept of impaired perfusion and forward flow recently has been modified to include the recognition of systemic venous congestion as a contributor, with direct and indirect mechanisms, including elevated renal venous pressure, reduced renal perfusion pressure, increased renal interstitial pressure, tubular dysfunction, splanchnic congestion, and neurohormonal and inflammatory activation. Treatment options beyond diuretics and vasoactive drugs remain limited and lack supportive evidence.
Collapse
Affiliation(s)
- Thida Tabucanon
- Kaufman Center for Heart Failure Treatment and Recovery, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH 44195, USA
| | - Wai Hong Wilson Tang
- Kaufman Center for Heart Failure Treatment and Recovery, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH 44195, USA; Department of Cardiovascular Medicine, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH 44195, USA.
| |
Collapse
|
18
|
Mayerhofer CCK, Kummen M, Holm K, Broch K, Awoyemi A, Vestad B, Storm-Larsen C, Seljeflot I, Ueland T, Bohov P, Berge RK, Svardal A, Gullestad L, Yndestad A, Aukrust P, Hov JR, Trøseid M. Low fibre intake is associated with gut microbiota alterations in chronic heart failure. ESC Heart Fail 2020; 7:456-466. [PMID: 31978943 PMCID: PMC7160496 DOI: 10.1002/ehf2.12596] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/15/2019] [Accepted: 11/27/2019] [Indexed: 12/30/2022] Open
Abstract
AIMS Recent reports have suggested that patients with heart failure (HF) have an altered gut microbiota composition; however, associations with diet remain largely uninvestigated. We aimed to explore differences in the gut microbiota between patients with HF with reduced ejection fraction and healthy controls, focusing on associations with diet and disease severity. METHODS AND RESULTS The microbiota composition of two cross-sectional cohorts (discovery, n = 40 and validation, n = 44) of patients with systolic HF and healthy controls (n = 266) was characterized by sequencing of the bacterial 16S rRNA gene. The overall microbial community (beta diversity) differed between patients with HF and healthy controls in both cohorts (P < 0.05). Patients with HF had shifts in the major bacterial phyla, resulting in a lower Firmicutes/Bacteroidetes (F/B) ratio than controls (P = 0.005). Patients reaching a clinical endpoint (listing for heart transplant or death) had lower bacterial richness and lower F/B ratio than controls (P < 0.01). Circulating levels of trimethylamine-N-oxide were associated with meat intake (P = 0.016), but not with gut microbiota alterations in HF. Low bacterial richness and low abundance of several genera in the Firmicutes phylum were associated with low fibre intake. CONCLUSIONS The gut microbiota in HF was characterized by decreased F/B ratio and reduced bacterial diversity associated with clinical outcome. The gut microbiota alterations in HF were partly related to low fibre intake, emphasizing the importance of diet as a covariate in future studies. Our data could provide a rationale for targeting the gut microbiota in HF with high-fibre diet.
Collapse
Affiliation(s)
- Cristiane C K Mayerhofer
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Martin Kummen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Norwegian PSC Research Center, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kristian Holm
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Norwegian PSC Research Center, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ayodeji Awoyemi
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Beate Vestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Christopher Storm-Larsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Norwegian PSC Research Center, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ingebjørg Seljeflot
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Pavol Bohov
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Rolf K Berge
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Asbjørn Svardal
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Lars Gullestad
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Section of Clinical Immunology and Infectious diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Johannes R Hov
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Norwegian PSC Research Center, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Section of Gastroenterology, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Section of Clinical Immunology and Infectious diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
19
|
Kostenko VA, Sitnikova MY, Skorodumova EA, Skorodumova EG, Fedorov AN, Pivovarova LP, Osipova IV, Ariskina OB, Rysev AV, Povzun AS. [Characteristic of infectious status in patients with acutely decompencated chronic heart failure and its impact on annual prognosis]. ACTA ACUST UNITED AC 2019; 59:56-62. [PMID: 31526362 DOI: 10.18087/cardio.2642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 09/16/2019] [Indexed: 11/18/2022]
Abstract
AIM The assessment of infectious status in patients with acutely decompensated chronic heart faiure (ADCHF) without evident signs of acute inflammatory stress and its impact on the 1 year prognosis. MATERIAL AND METHODS Totally, 65 patients with ADCHF of ischemic origin investigated, age 67,3±2,3 y.o. All patients were taken markers of phagocytosis and inflammatory stress as well as antibodies to Streptococcus, Cytomegalovirus (CMV), Epstein-Barr virus (VEB), Candida albicans, Toxoplasma gondii, Aspergillus, Mycoplasma hominis and pneumonia and also level of lipopolysaccharids (LPS) of gram-negative bacteriae. RESULTS More often LPS of gram-negative bacteriae were revealed in patients with ADCHF and further in decreasing order - antibodies to CMV, VEB, Streptococcus, Candida, Aspergillus and LPS. All patients have been infected by at least 2 pathogens, more than 90 % of them had 3 ones or more. Mortality in first 12 months observation correlated with quantity of patient`s pathogenic patterns (r=0,52, p=0,004). Dependency of one-year mortality from degree of viral-bacterial mixt contamination was almost linear. CMV was a monopathogen with strongest correlation with mortality (r=0,39, p=0,001). In patients with more significant infection bigger rate of re-hospitalizations about new ADCHF correlated with number of pathogens was observed (r=0,61, p=0,001). CONCLUSION Chronic latent infection with a significant number of pathogens is characteristic of patients with low-ejection ADCHF of ischemic genesis with a significant number of pathogens: more than 90 % of patients had three or more. The most common exogenous pathogens in the study sample of patients with chronic obstructive heart failure were CMV, EBV, and hemolytic streptococcus, of the potentially endogenous ones, gram-negative intestinal bacteria. The number of infectious agents in patients with chronic obstructive heart failure has a direct correlation with deaths and re-admission to hospital with total heart failure within 1 year after discharge from the hospital.
Collapse
Affiliation(s)
- V A Kostenko
- Research Institute of Emergency medicine named after I. I. Janelidze
| | | | - E A Skorodumova
- Research Institute of Emergency medicine named after I. I. Janelidze
| | - E G Skorodumova
- Research Institute of Emergency medicine named after I. I. Janelidze
| | - A N Fedorov
- Research Institute of Emergency medicine named after I. I. Janelidze
| | - L P Pivovarova
- Research Institute of Emergency medicine named after I. I. Janelidze
| | - I V Osipova
- Research Institute of Emergency medicine named after I. I. Janelidze
| | - O B Ariskina
- Research Institute of Emergency medicine named after I. I. Janelidze
| | - A V Rysev
- Research Institute of Emergency medicine named after I. I. Janelidze
| | - A S Povzun
- Research Institute of Emergency medicine named after I. I. Janelidze
| |
Collapse
|
20
|
Virzì GM, Breglia A, Ankawi G, Bolin C, de Cal M, Cianci V, Vescovo G, Ronco C. Plasma Lipopolysaccharide Concentrations in Cardiorenal Syndrome Type 1. Cardiorenal Med 2019; 9:308-315. [PMID: 31238313 DOI: 10.1159/000500480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cardiorenal syndrome (CRS) type 1 is characterized by a rapid worsening of cardiac function that leads to acute kidney injury (AKI). This study evaluated the role of lipopolysaccharide (LPS) in the development of AKI in patients with acute heart failure (AHF) and its relationship with renal parameters, to enable a better comprehension of the pathophysiology of CRS type 1. METHODS We enrolled 32 AHF patients, 15 of whom were classified as having CRS type 1. Eight of these 15 exhibited AKI at the time of admission (caused by AHF) and the other 7 developed AKI during their stay in hospital (in the first 48 h). We evaluated the plasmatic LPS concentrations as well as conventional (serum creatinine [sCr] and urea) and unconventional (neutrophil gelatinase-associated lipocalin [NGAL] and cystatin C) renal markers. RESULTS LPS levels were significantly higher in the CRS type 1 patients. No significant difference in LPS level was found in patients who were admitted with AKI and those developed AKI in hospital, but there was a tendency towards a higher level of LPS in CRS type 1 patients admitted with AKI. The LPS concentrations at admission were similar in CRS type 1 survivors (n = 12) and nonsurvivors (n = 3) (p = 0.22). We observed a positive correlation between LPS level and NGAL, Scr at admission and peak Scr during hospitalization and urea at admission. CONCLUSION CRS type 1 patients present with an increased level of LPS and there is a direct correlation between LPS and renal parameters. This pilot research is the first study to explore the premise of LPS as novel pathophysiological factor in CRS type 1.
Collapse
Affiliation(s)
- Grazia Maria Virzì
- IRRIV-International Renal Research Institute Vicenza, Vicenza, Italy, .,Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, Vicenza, Italy,
| | - Andrea Breglia
- IRRIV-International Renal Research Institute Vicenza, Vicenza, Italy.,Emergency Department of Arzignano Hospital, Arzignano, Italy
| | - Ghada Ankawi
- IRRIV-International Renal Research Institute Vicenza, Vicenza, Italy.,Department of Internal Medicine and Nephrology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Chiara Bolin
- Internal Medicine, San Bortolo Hospital, Vicenza, Italy
| | - Massimo de Cal
- IRRIV-International Renal Research Institute Vicenza, Vicenza, Italy.,Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, Vicenza, Italy
| | - Vito Cianci
- Emergency Department of Arzignano Hospital, Arzignano, Italy
| | - Giorgio Vescovo
- Internal Medicine, San Bortolo Hospital, Vicenza, Italy.,Internal Medicine Unit, Sant'Antonio Hospital Padua, Padua, Italy
| | - Claudio Ronco
- IRRIV-International Renal Research Institute Vicenza, Vicenza, Italy.,Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, Vicenza, Italy.,Nephrology, Department of Medicine, University of Padua, Padua, Italy
| |
Collapse
|
21
|
Al-Rubaye H, Perfetti G, Kaski JC. The Role of Microbiota in Cardiovascular Risk: Focus on Trimethylamine Oxide. Curr Probl Cardiol 2019; 44:182-196. [DOI: 10.1016/j.cpcardiol.2018.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 06/07/2018] [Indexed: 02/08/2023]
|
22
|
Lipopolysaccharide in systemic circulation induces activation of inflammatory response and oxidative stress in cardiorenal syndrome type 1. J Nephrol 2019; 32:803-810. [DOI: 10.1007/s40620-019-00613-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/15/2019] [Indexed: 01/21/2023]
|
23
|
Ferrini A, Stevens MM, Sattler S, Rosenthal N. Toward Regeneration of the Heart: Bioengineering Strategies for Immunomodulation. Front Cardiovasc Med 2019; 6:26. [PMID: 30949485 PMCID: PMC6437044 DOI: 10.3389/fcvm.2019.00026] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/26/2019] [Indexed: 01/10/2023] Open
Abstract
Myocardial Infarction (MI) is the most common cardiovascular disease. An average-sized MI causes the loss of up to 1 billion cardiomyocytes and the adult heart lacks the capacity to replace them. Although post-MI treatment has dramatically improved survival rates over the last few decades, more than 20% of patients affected by MI will subsequently develop heart failure (HF), an incurable condition where the contracting myocardium is transformed into an akinetic, fibrotic scar, unable to meet the body's need for blood supply. Excessive inflammation and persistent immune auto-reactivity have been suggested to contribute to post-MI tissue damage and exacerbate HF development. Two newly emerging fields of biomedical research, immunomodulatory therapies and cardiac bioengineering, provide potential options to target the causative mechanisms underlying HF development. Combining these two fields to develop biomaterials for delivery of immunomodulatory bioactive molecules holds great promise for HF therapy. Specifically, minimally invasive delivery of injectable hydrogels, loaded with bioactive factors with angiogenic, proliferative, anti-apoptotic and immunomodulatory functions, is a promising route for influencing the cascade of immune events post-MI, preventing adverse left ventricular remodeling, and offering protection from early inflammation to fibrosis. Here we provide an updated overview on the main injectable hydrogel systems and bioactive factors that have been tested in animal models with promising results and discuss the challenges to be addressed for accelerating the development of these novel therapeutic strategies.
Collapse
Affiliation(s)
- Arianna Ferrini
- Department of Materials, Imperial College London, London, United Kingdom,National Heart and Lung Institute and BHF Centre for Research Excellence, Imperial College London, London, United Kingdom
| | - Molly M. Stevens
- Department of Materials, Imperial College London, London, United Kingdom,Department of Bioengineering, Imperial College London, London, United Kingdom,Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Susanne Sattler
- National Heart and Lung Institute and BHF Centre for Research Excellence, Imperial College London, London, United Kingdom
| | - Nadia Rosenthal
- National Heart and Lung Institute and BHF Centre for Research Excellence, Imperial College London, London, United Kingdom,The Jackson Laboratory, Bar Harbor, ME, United States,*Correspondence: Nadia Rosenthal
| |
Collapse
|
24
|
Samokhvalov V, Jamieson KL, Darwesh AM, Keshavarz-Bahaghighat H, Lee TYT, Edin M, Lih F, Zeldin DC, Seubert JM. Deficiency of Soluble Epoxide Hydrolase Protects Cardiac Function Impaired by LPS-Induced Acute Inflammation. Front Pharmacol 2019; 9:1572. [PMID: 30692927 PMCID: PMC6339940 DOI: 10.3389/fphar.2018.01572] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/24/2018] [Indexed: 12/14/2022] Open
Abstract
Lipopolysaccharide (LPS) is a bacterial wall endotoxin producing many pathophysiological conditions including myocardial inflammation leading to cardiotoxicity. Linoleic acid (18:2n6, LA) is an essential n-6 PUFA which is converted to arachidonic acid (20:4n6, AA) by desaturation and elongation via enzyme systems within the body. Biological transformation of PUFA through CYP-mediated hydroxylation, epoxidation, and allylic oxidation produces lipid mediators, which may be subsequently hydrolyzed to corresponding diol metabolites by soluble epoxide hydrolase (sEH). In the current study, we investigate whether inhibition of sEH, which alters the PUFA metabolite profile, can influence LPS induced cardiotoxicity and mitochondrial function. Our data demonstrate that deletion of soluble epoxide hydrolase provides protective effects against LPS-induced cardiotoxicity by maintaining mitochondrial function. There was a marked alteration in the cardiac metabolite profile with notable increases in sEH-derived vicinal diols, 9,10- and 12,13-dihydroxyoctadecenoic acid (DiHOME) in WT hearts following LPS administration, which was absent in sEH null mice. We found that DiHOMEs triggered pronounced mitochondrial structural abnormalities, which also contributed to the development of extensive mitochondrial dysfunction in cardiac cells. Accumulation of DiHOMEs may represent an intermediate mechanism through which LPS-induced acute inflammation triggers deleterious alterations in the myocardium in vivo and cardiac cells in vitro. This study reveals novel research exploring the contribution of DiHOMEs in the progression of adverse inflammatory responses toward cardiac function in vitro and in vivo.
Collapse
Affiliation(s)
- Victor Samokhvalov
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - K Lockhart Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Ahmed M Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | | | - Tim Y T Lee
- Department of Pharmacology, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Matthew Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Fred Lih
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Department of Pharmacology, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
25
|
Sandstedt J, Sandstedt M, Lundqvist A, Jansson M, Sopasakis VR, Jeppsson A, Hultén LM. Human cardiac fibroblasts isolated from patients with severe heart failure are immune-competent cells mediating an inflammatory response. Cytokine 2018; 113:319-325. [PMID: 30360948 DOI: 10.1016/j.cyto.2018.09.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/24/2018] [Accepted: 09/29/2018] [Indexed: 01/26/2023]
Abstract
This study was aimed to elucidate the immunoregulatory properties of human cardiac fibroblasts cultured under pro-inflammatory and hypoxic conditions. Human heart tissue for isolating cardiac cells is generally hard to obtain, particularly from all four chambers of the same heart. Since different parts of the heart have different functions and therefore may have different immunoregulatory properties, ability to analyse cells from all chambers allows for a unique and comprehensive investigation. Cells were isolated from all four chambers of the heart from patients undergoing cardiac transplantation surgery due to severe chronic heart failure (CHF) (n = 6). Cells isolated from one donor heart, were used for comparison with the experimental group. Primary cultured human cardiac fibroblasts were treated with Lipopolysaccharide (LPS) to induce an inflammatory response. Cells were also subjected to hypoxia. To determine immunoregulatory properties of the cells, cytokine and chemokine profiles were determined using multiplex ELISA. RESULTS: On average, the fibroblasts population constituted approximately 90% of the expanded non-myocytes. Levels of cytokines and chemokines were markedly increased in human cardiac fibroblasts cultured under inflammatory conditions, with a similar response in fibroblasts from all compartments of the heart. Unexpectedly, hypoxia did not further augment cytokine and chemokine secretion. In conclusion, human cardiac fibroblasts are a robust source of pro-inflammatory mediators in the failing heart, independent of hypoxia, and might play a critical role in inflammation associated with the pathogenesis of CHF.
Collapse
Affiliation(s)
- Joakim Sandstedt
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg and Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Mikael Sandstedt
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg and Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Annika Lundqvist
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg and Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Märta Jansson
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg and Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Victoria Rotter Sopasakis
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg and Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anders Jeppsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Lillemor Mattsson Hultén
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg and Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
26
|
Mayerhofer CCK, Awoyemi AO, Moscavitch SD, Lappegård KT, Hov JR, Aukrust P, Hovland A, Lorenzo A, Halvorsen S, Seljeflot I, Gullestad L, Trøseid M, Broch K. Design of the GutHeart-targeting gut microbiota to treat heart failure-trial: a Phase II, randomized clinical trial. ESC Heart Fail 2018; 5:977-984. [PMID: 30088346 PMCID: PMC6165929 DOI: 10.1002/ehf2.12332] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 05/21/2018] [Accepted: 06/14/2018] [Indexed: 12/20/2022] Open
Abstract
Aims Heart failure (HF) is a multifactorial disease. Current treatments target only a fraction of the putative pathophysiological pathways. In patients with HF, reduced cardiac output and congestion cause increased gut wall permeability. It has been suggested that leakage of microbial products is detrimental to the heart, at least partly through activation of systemic inflammatory pathways, which again could promote gut leakage. Whether manipulating the gut microbiota can improve cardiac function in patients with HF remains unknown. We aim to evaluate the effect of drugs targeting the gut microbiota on left ventricular function, quality of life, and functional capacity, as well as on markers of gut leakage and inflammation, in stable patients with HF with reduced ejection fraction. Methods and results GutHeart is a randomized, open‐label, controlled trial. Four centres will randomize 150 patients with stable HF and a left ventricular ejection fraction <40% to receive the antibiotic rifaximin, the probiotic yeast Saccharomyces boulardii (ATCC 74012), or no treatment (control) in a 1:1:1 fashion. Treatment will last for 3 months. The primary endpoint is baseline‐adjusted left ventricular ejection fraction as measured by echocardiography after 3 months. A further follow‐up 6 months after randomization will be undertaken. Conclusions This trial is likely to give new insights into important disease processes involving the gut microbiota in HF patients, hereby leading to new potential therapeutic strategies to prevent and down‐regulate the inflammation seen in these patients.
Collapse
Affiliation(s)
- Cristiane C K Mayerhofer
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ayodeji O Awoyemi
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway.,Department of Cardiology, Oslo University Hospital, Ullevål, Postboks 4956 Nydalen, 0424, Oslo, Norway
| | - Samuel D Moscavitch
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Knut Tore Lappegård
- Division of Internal Medicine, Nordlandssykehuset, Bodø, Norway.,Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway
| | - Johannes R Hov
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Norwegian PSC Research Center and Section of Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammation Medicine and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Anders Hovland
- Division of Internal Medicine, Nordlandssykehuset, Bodø, Norway.,Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway
| | - Andrea Lorenzo
- Instituto Nacional de Cardiologia, Rio de Janeiro, Brazil
| | - Sigrun Halvorsen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Cardiology, Oslo University Hospital, Ullevål, Postboks 4956 Nydalen, 0424, Oslo, Norway
| | - Ingebjørg Seljeflot
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Lars Gullestad
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
27
|
Cosola C, Sabatino A, di Bari I, Fiaccadori E, Gesualdo L. Nutrients, Nutraceuticals, and Xenobiotics Affecting Renal Health. Nutrients 2018; 10:nu10070808. [PMID: 29937486 PMCID: PMC6073437 DOI: 10.3390/nu10070808] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) affects 8–16% of the population worldwide. In developed countries, the most important risk factors for CKD are diabetes, hypertension, and obesity, calling into question the importance of educating and acting on lifestyles and nutrition. A balanced diet and supplementation can indeed support the maintenance of a general health status, including preservation of renal function, and can help to manage and curb the main risk factors for renal damage. While the concept of protein and salt restriction in nephrology is historically acknowledged, the role of some nutrients in renal health and the importance of nutrition as a preventative measure for renal care are less known. In this narrative review, we provide an overview of the demonstrated and potential actions of some selected nutrients, nutraceuticals, and xenobiotics on renal health and function. The direct and indirect effects of fiber, protein, fatty acids, curcumin, steviol glycosides, green tea, coffee, nitrates, nitrites, and alcohol on kidney health are reviewed here. In view of functional and personalized nutrition, understanding the renal and systemic effects of dietary components is essential since many chronic conditions, including CKD, are related to systemic dysfunctions such as chronic low-grade inflammation.
Collapse
Affiliation(s)
- Carmela Cosola
- Department of Emergency and Organ Transplantation-Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, 70124 Bari, Italy.
| | - Alice Sabatino
- Department of Medicine and Surgery, Parma University Medical School, 43126 Parma, Italy.
| | - Ighli di Bari
- Department of Emergency and Organ Transplantation-Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, 70124 Bari, Italy.
| | - Enrico Fiaccadori
- Department of Medicine and Surgery, Parma University Medical School, 43126 Parma, Italy.
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation-Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, 70124 Bari, Italy.
| |
Collapse
|
28
|
Yang J, Lim SY, Ko YS, Lee HY, Oh SW, Kim MG, Cho WY, Jo SK. Intestinal barrier disruption and dysregulated mucosal immunity contribute to kidney fibrosis in chronic kidney disease. Nephrol Dial Transplant 2018; 34:419-428. [DOI: 10.1093/ndt/gfy172] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/08/2018] [Indexed: 12/19/2022] Open
Affiliation(s)
- Jihyun Yang
- Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Sung Yoon Lim
- Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Yoon Sook Ko
- Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Hee Young Lee
- Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Se Won Oh
- Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Myung Gyu Kim
- Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Won Yong Cho
- Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Sang Kyung Jo
- Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| |
Collapse
|
29
|
Black AP, Anjos JS, Cardozo L, Carmo FL, Dolenga CJ, Nakao LS, de Carvalho Ferreira D, Rosado A, Carraro Eduardo JC, Mafra D. Does Low-Protein Diet Influence the Uremic Toxin Serum Levels From the Gut Microbiota in Nondialysis Chronic Kidney Disease Patients? J Ren Nutr 2018; 28:208-214. [PMID: 29439931 DOI: 10.1053/j.jrn.2017.11.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/23/2017] [Accepted: 11/28/2017] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES To evaluate the effects of low-protein diet (LPD) on uremic toxins and the gut microbiota profile in nondialysis chronic kidney disease (CKD) patients. DESIGN AND METHODS Longitudinal study with 30 nondialysis CKD patients (stage 3-4) undergoing LPD for 6 months. Adherence to the diet was evaluated based on the calculation of protein equivalent of nitrogen appearance from the 24-hour urine analysis. Good adherence to LPD was considered when protein intake was from 90% to 110% of the prescribed amount (0.6 g/kg/day). Food intake was analyzed by the 24-hour recall method. The anthropometric, biochemical and lipid profile parameters were measured according to standard methods. Uremic toxin serum levels (indoxyl sulfate, p-cresyl sulfate, indole-3-acetic acid) were obtained by reversed-phase high-performance liquid chromatography (RP-HPLC). Fecal samples were collected to evaluate the gut microbiota profile through polymerase chain reaction and denaturing gradient gel electrophoresis. Statistical analysis was performed by the SPSS 23.0 program software. RESULTS Patients who adhered to the diet (n = 14) (0.7 ± 0.2 g/kg/day) presented an improvement in renal function (nonsignificant) and reduction in total and low-density lipoprotein cholesterol (183.9 ± 48.5-155.7 ± 37.2 mg/dL, P = .01; 99.4 ± 41.3-76.4 ± 33.2 mg/dL, P = .01, respectively). After 6 months of nutricional intervention, p-cresyl sulfate serum levels were reduced significantly in patients who adhered to the LPD (19.3 [9.6-24.7] to 15.5 [9.8-24.1] mg/L, P = .03), and in contrast, the levels were increased in patients who did not adhere (13.9 [8.0-24.8] to 24.3 [8.1-39.2] mg/L, P = .004). In addition, using the denaturing gradient gel electrophoresis technique, it was observed change in the intestinal microbiota profile after LPD intervention in both groups, and the number of bands was positively associated with protein intake (r = 0.44, P = .04). CONCLUSION LPD seems be a good strategy to reduce the uremic toxins production by the gut microbiota in nondialysis CKD patients.
Collapse
Affiliation(s)
- Ana Paula Black
- Post Graduation Program in Medical Sciences, Fluminense Federal University (UFF), Niterói-RJ, Brazil.
| | - Juliana S Anjos
- Post Graduation Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói-RJ, Brazil
| | - Ludmila Cardozo
- Post Graduation Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói-RJ, Brazil
| | - Flávia L Carmo
- Institute of Microbiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Carla J Dolenga
- Basic Pathology Department, Federal University of Paraná (UFPR), Curitiba-PR, Brazil
| | - Lia S Nakao
- Basic Pathology Department, Federal University of Paraná (UFPR), Curitiba-PR, Brazil
| | | | - Alexandre Rosado
- Institute of Microbiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | | - Denise Mafra
- Post Graduation Program in Medical Sciences, Fluminense Federal University (UFF), Niterói-RJ, Brazil; Post Graduation Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói-RJ, Brazil
| |
Collapse
|
30
|
Visceral Congestion in Heart Failure: Right Ventricular Dysfunction, Splanchnic Hemodynamics, and the Intestinal Microenvironment. Curr Heart Fail Rep 2018; 14:519-528. [PMID: 29075956 DOI: 10.1007/s11897-017-0370-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Visceral venous congestion of the gut may play a key role in the pathogenesis of right-sided heart failure (HF) and cardiorenal syndromes. Here, we review the role of right ventricular (RV) dysfunction, visceral congestion, splanchnic hemodynamics, and the intestinal microenvironment in the setting of right-sided HF. We review recent literature on this topic, outline possible mechanisms of disease pathogenesis, and discuss potential therapeutics. RECENT FINDINGS There are several mechanisms linking RV-gut interactions via visceral venous congestion which could result in (1) hypoxia and acidosis in enterocytes, which may lead to enhanced sodium-hydrogen exchanger 3 (NHE3) expression with increased sodium and fluid retention; (2) decreased luminal pH in the intestines, which could lead to alteration of the gut microbiome which could increase gut permeability and inflammation; (3) alteration of renal hemodynamics with triggering of the cardiorenal syndrome; and (4) altered phosphate metabolism resulting in increased pulmonary artery stiffening, thereby increasing RV afterload. A wide variety of therapeutic interventions that act on the RV, pulmonary vasculature, intestinal microenvironment, and the kidney could alter these pathways and should be tested in patients with right-sided HF. The RV-gut axis is an important aspect of HF pathogenesis that deserves more attention. Modulation of the pathways interconnecting the right heart, visceral congestion, and the intestinal microenvironment could be a novel avenue of intervention for right-sided HF.
Collapse
|
31
|
Jiménez JA, Peterson CT, Mills PJ. Neuroimmune Mechanisms of Depression in Adults with Heart Failure. Methods Mol Biol 2018; 1781:145-169. [PMID: 29705847 DOI: 10.1007/978-1-4939-7828-1_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Heart failure (HF) is a major and costly public health concern, and its prognosis is grim-with high hospitalization and mortality rates. HF affects millions of individuals across the world, and this condition is expected to become "the epidemic" of the twenty-first century (Jessup et al., 2016). It is well documented that individuals with HF experience disproportionately high rates of depression and that those who are depressed have worse clinical outcomes than their nondepressed counterparts. The purpose of this chapter is to introduce the reader to the study of depression in HF, and how psychoneuroimmunologic principles have been applied to further elucidate mechanisms (i.e., neurohormonal and cytokine activation) linking these comorbid disorders.
Collapse
Affiliation(s)
- Jessica A Jiménez
- Department of Psychology, College of Letters and Sciences, National University, La Jolla, CA, USA.
| | - Christine Tara Peterson
- Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Paul J Mills
- Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
32
|
Grant C, Harrison L, Hoad C, Marciani L, Cox E, Buchanan C, Costigan C, Francis S, Lai KB, Szeto CC, Gowland P, McIntyre C. Endotoxemia in Peritoneal Dialysis Patients: A Pilot Study to Examine the Role of Intestinal Perfusion and Congestion. Perit Dial Int 2017; 37:111-115. [PMID: 28153967 DOI: 10.3747/pdi.2016.00079] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Endotoxemia is common in advanced chronic kidney disease and is particularly severe in those receiving dialysis. In hemodialysis patients, translocation from the bowel occurs as a consequence of recurrent circulatory stress leading to a reduction in circulating splanchnic volume and increased intestinal permeability. Peritoneal dialysis (PD) patients are often volume expanded and have continuous direct immersion of bowel in fluid; these may also be important factors in endotoxin translocation and would suggest different therapeutic strategies to improve it. The mechanisms leading to endotoxemia have never been specifically studied in PD. In this study, 17 subjects (8 PD patients, 9 healthy controls) underwent detailed gastrointestinal and cardiac magnetic resonance imaging during fasted and fed states. Gross splanchnic perfusion was assessed by quantification of superior mesenteric artery flow. Magnetic resonance imaging findings were correlated to endotoxemia, markers of hydration status and cardiac structure and function.
Collapse
Affiliation(s)
- Claire Grant
- University of Western Ontario, Kidney Clinical Research Unit, London, ON, Canada
| | - Laura Harrison
- Royal Derby Hospital, Department of Renal Medicine, Derby, UK
| | - Caroline Hoad
- University of Nottingham, Sir Peter Mansfield Imaging Centre, Nottingham, UK
| | - Luca Marciani
- University of Nottingham, Nottingham Digestive Diseases Centre, Nottingham, UK
| | - Eleanor Cox
- University of Nottingham, Sir Peter Mansfield Imaging Centre, Nottingham, UK
| | - Charlotte Buchanan
- University of Nottingham, Sir Peter Mansfield Imaging Centre, Nottingham, UK
| | - Carolyn Costigan
- University of Nottingham, Sir Peter Mansfield Imaging Centre, Nottingham, UK
| | - Susan Francis
- University of Nottingham, Sir Peter Mansfield Imaging Centre, Nottingham, UK
| | - Ka-Bik Lai
- The Chinese University of Hong Kong, Department of Medicine & Therapeutics, Sha Tin, Hong Kong
| | - Cheuk-Chun Szeto
- The Chinese University of Hong Kong, Department of Medicine & Therapeutics, Sha Tin, Hong Kong
| | - Penny Gowland
- University of Nottingham, Sir Peter Mansfield Imaging Centre, Nottingham, UK
| | - Christopher McIntyre
- University of Western Ontario, Kidney Clinical Research Unit, London, ON, Canada
| |
Collapse
|
33
|
Chou JA, Kalantar-Zadeh K, Mathew AT. A brief review of intradialytic hypotension with a focus on survival. Semin Dial 2017; 30:473-480. [PMID: 28661565 PMCID: PMC5738929 DOI: 10.1111/sdi.12627] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Intradialytic hypotension (IDH), a common complication of ultrafiltration during hemodialysis therapy, is associated with high mortality and morbidity. IDH, defined as a nadir systolic blood pressure of less than 90 mm Hg on more than 30% of treatments, is a relevant definition and is correlated with mortality. Risk factors for IDH include patient demographics, anti-hypertensive medication use, larger interdialytic weight gain, and dialysis prescription features as dialysate sodium, high ultrafiltration rate, and dialysate temperature. A high frequency of IDH events carries a substantial death risk. An ultrafiltration rate >10 mL/h/kg, and even more so >13 mL/h/kg, is highly predictive of cardiovascular and all-cause mortality. Evidence suggests that IDH causes acute reversible segmental myocardial hypoperfusion and contractile dysfunction (myocardial stunning), which can result in long-term loss of myocardial contractility, leading to premature death. IDH also has negative end-organ effects on the brain and gut, contributing to mortality through stroke, and endotoxin translocation with associated inflammation and protein-energy wasting. Given strong association of IDH and dialysis mortality, a paradigm shift to its approach is urgently needed. Randomized controlled trials are required to prospectively test drugs and monitoring devices which may reduce IDH.
Collapse
Affiliation(s)
- Jason A Chou
- Division of Nephrology and Hypertension, Harold Simmons Center for Kidney Disease Research and Epidemiology, Los Angeles, CA, USA
| | - Kamyar Kalantar-Zadeh
- Division of Nephrology and Hypertension, Harold Simmons Center for Kidney Disease Research and Epidemiology, Los Angeles, CA, USA
- Fielding School of Public Health at UCLA, Los Angeles, CA, USA
- Los Angeles Biomedical Research Institute at Harbor-UCLA, Torrance, CA, USA
| | - Anna T Mathew
- Division of Nephrology, Northwell Health, Great Neck, NY, USA
| |
Collapse
|
34
|
de Melo LGP, Nunes SOV, Anderson G, Vargas HO, Barbosa DS, Galecki P, Carvalho AF, Maes M. Shared metabolic and immune-inflammatory, oxidative and nitrosative stress pathways in the metabolic syndrome and mood disorders. Prog Neuropsychopharmacol Biol Psychiatry 2017; 78:34-50. [PMID: 28438472 DOI: 10.1016/j.pnpbp.2017.04.027] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/27/2017] [Accepted: 04/08/2017] [Indexed: 02/08/2023]
Abstract
This review examines the shared immune-inflammatory, oxidative and nitrosative stress (IO&NS) and metabolic pathways underpinning metabolic syndrome (MetS), bipolar disorder (BD) and major depressive disorder (MDD). Shared pathways in both MetS and mood disorders are low grade inflammation, including increased levels of pro-inflammatory cytokines and acute phase proteins, increased lipid peroxidation with formation of malondialdehyde and oxidized low density lipoprotein cholesterol (LDL-c), hypernitrosylation, lowered levels of antioxidants, most importantly zinc and paraoxonase (PON1), increased bacterial translocation (leaky gut), increased atherogenic index of plasma and Castelli risk indices; and reduced levels of high-density lipoprotein (HDL-c) cholesterol. Insulin resistance is probably not a major factor associated with mood disorders. Given the high levels of IO&NS and metabolic dysregulation in BD and MDD and the high comorbidity with the atherogenic components of the MetS, mood disorders should be viewed as systemic neuro-IO&NS-metabolic disorders. The IO&NS-metabolic biomarkers may have prognostic value and may contribute to the development of novel treatments targeting neuro-immune, neuro-oxidative and neuro-nitrosative pathways.
Collapse
Affiliation(s)
- Luiz Gustavo Piccoli de Melo
- Department of Clinical Medicine, Londrina State University (UEL), Health Sciences Centre, Londrina, Paraná, Brazil; Center of Approach and Treatment for Smokers, University Hospital, Londrina State University, University Campus, Londrina, Paraná, Brazil; Health Sciences Graduation Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Sandra Odebrecht Vargas Nunes
- Department of Clinical Medicine, Londrina State University (UEL), Health Sciences Centre, Londrina, Paraná, Brazil; Center of Approach and Treatment for Smokers, University Hospital, Londrina State University, University Campus, Londrina, Paraná, Brazil; Health Sciences Graduation Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | | | - Heber Odebrecht Vargas
- Department of Clinical Medicine, Londrina State University (UEL), Health Sciences Centre, Londrina, Paraná, Brazil; Center of Approach and Treatment for Smokers, University Hospital, Londrina State University, University Campus, Londrina, Paraná, Brazil; Health Sciences Graduation Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Décio Sabbattini Barbosa
- Health Sciences Graduation Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil; Department of Clinical and Toxicological Analysis, State University of Londrina, Londrina, Paraná, Brazil
| | - Piotr Galecki
- Department of Adult Psychiatry, University of Lodz, Lodz, Poland
| | - André F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Michael Maes
- Health Sciences Graduation Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil; Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Plovdiv University, Plovdiv, Bulgaria; Revitalis, Waalre, The Netherlands; Impact Strategic Research Center, Deakin University, Geelong, Australia.
| |
Collapse
|
35
|
Ryzhov S, Matafonov A, Galindo CL, Zhang Q, Tran TL, Lenihan DJ, Lenneman CG, Feoktistov I, Sawyer DB. ERBB signaling attenuates proinflammatory activation of nonclassical monocytes. Am J Physiol Heart Circ Physiol 2017; 312:H907-H918. [PMID: 28235789 DOI: 10.1152/ajpheart.00486.2016] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 02/22/2017] [Accepted: 02/22/2017] [Indexed: 01/01/2023]
Abstract
Immune activation in chronic systolic heart failure (HF) correlates with disease severity and prognosis. Recombinant neuregulin-1 (rNRG-1) is being developed as a possible therapy for HF, based on the activation of ERBB receptors in cardiac cells. Work in animal models of HF led us to hypothesize that there may be direct effects of NRG-1 on immune system activation and inflammation. We investigated the expression of ERBB receptors and the effect of rNRG-1 isoform glial growth factor 2 (GGF2) in subpopulations of peripheral blood mononuclear cells (PB MNCs) in subjects with HF. We found that human monocytes express both ERBB2 and ERBB3 receptors, with high interindividual variability among subjects. Monocyte surface ERBB3 and TNF-α mRNA expression were inversely correlated in subjects with HF but not in human subjects without HF. GGF2 activation of ERBB signaling ex vivo inhibited LPS-induced TNF-α production, specifically in the CD14lowCD16+ population of monocytes in a phosphoinositide 3-kinase-dependent manner. GGF2 suppression of TNF-α correlated directly with the expression of ERBB3. In vivo, a single dose of intravenous GGF2 reduced TNF-α expression in PB MNCs of HF subjects participating in a phase I safety study of GGF2. These results support a role for ERBB3 signaling in the regulation of TNF-α production from CD14lowCD16+ monocytes and a need for further investigation into the clinical significance of NRG-1/ERBB signaling as a modulator of immune system function.NEW & NOTEWORTHY This study identified a novel role of neuregulin-1 (NRG-1)/ERBB signaling in the control of proinflammatory activation of monocytes. These results further improve our fundamental understanding of cardioprotective effects of NRG-1 in patients with heart failure.
Collapse
Affiliation(s)
- Sergey Ryzhov
- Maine Medical Center Research Institute, Scarborough, Maine
| | - Anton Matafonov
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee.,Department of Bioengineering and Organic Chemistry, Tomsk Polytechnic University, Tomsk, Russia
| | - Cristi L Galindo
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Qinkun Zhang
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Truc-Linh Tran
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Daniel J Lenihan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Igor Feoktistov
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas B Sawyer
- Maine Medical Center Research Institute, Scarborough, Maine; .,Maine Medical Center, Portland, Maine
| |
Collapse
|
36
|
Virzì GM, Clementi A, Brocca A, Ronco C. Endotoxin Effects on Cardiac and Renal Functions and Cardiorenal Syndromes. Blood Purif 2017; 44:314-326. [PMID: 29161706 DOI: 10.1159/000480424] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 08/10/2017] [Indexed: 01/03/2023]
Abstract
Gram-negative sepsis is a major cause of morbidity and mortality in critical ill patients. Recent findings in molecular biology and in signaling pathways have enhanced our understanding of its pathogenesis and opened up opportunities of innovative therapeutic approaches. Endotoxin plays a pivotal role in the pathogenesis of multi-organ dysfunction in the setting of gram-negative sepsis. Indeed, heart and kidney impairments seem to be induced by the release of circulating pro-inflammatory and pro-apoptotic mediators triggered by endotoxin interaction with immune cells. These molecules are responsible for cellular apoptosis, autophagy, cell cycle arrest, and microRNAs activation. Therefore, the early identification of sepsis-associated acute kidney injury and heart dysfunction may improve the patient clinical outcome. In this report, we will consider the role of endotoxin in the pathogenesis of sepsis, its effects on both cardiac and renal functions, and the interactions between these 2 systems in the setting of cardiorenal syndromes (CRS), particularly in CRS type 5. Finally, we will discuss the possible role of extracorporeal therapies in reducing endotoxin levels.
Collapse
Affiliation(s)
- Grazia Maria Virzì
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, Agrigento, Italy
| | | | | | | |
Collapse
|
37
|
Abstract
Recent years have brought interesting insights into the human gut microbiota and have highlighted its increasingly recognized impact on cardiovascular (CV) diseases, including heart failure (HF). Changes in composition of gut microbiota, called dysbiosis, can trigger systemic inflammation, which is known to be involved in the pathophysiology of HF. Trimethylamine N-oxide (TMAO), which is derived from gut microbiota metabolites of specific dietary nutrients, has emerged as a key contributor to cardiovascular disease pathogenesis. Elevated TMAO levels have been reported to be associated with poor outcomes in patients with both HF and chronic kidney disease (CKD). Dysbiosis of gut microbiota can contribute to higher levels of TMAO and the generation of uremic toxins, progressing to both HF and CKD. Therefore, this bidirectional relationship between HF and CKD through gut microbiota may be a novel therapeutic target for the cardiorenal syndrome. However, the mechanisms by which gut microbiota could influence the development of heart failure are still unknown, and there are still some questions regarding the causative effects of TMAO and the underlying mechanistic link that explains how TMAO might directly or indirectly promote CV diseases including HF. Further studies are warranted to clarify the function of TMAO on the pathophysiology of cardiorenal syndrome and the handling of TMAO levels by the kidneys.
Collapse
|
38
|
|
39
|
Zhang X, Zhang X, Xiong Y, Xu C, Liu X, Lin J, Mu G, Xu S, Liu W. Sarcolemmal ATP-sensitive potassium channel protects cardiac myocytes against lipopolysaccharide-induced apoptosis. Int J Mol Med 2016; 38:758-66. [PMID: 27430376 PMCID: PMC4990318 DOI: 10.3892/ijmm.2016.2664] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 06/21/2016] [Indexed: 02/01/2023] Open
Abstract
The sarcolemmal ATP-sensitive K+ (sarcKATP) channel plays a cardioprotective role during stress. However, the role of the sarcKATP channel in the apoptosis of cardiomyocytes and association with mitochondrial calcium remains unclear. For this purpose, we developed a model of LPS-induced sepsis in neonatal rat cardiomyocytes (NRCs). The TUNEL assay was performed in order to detect the apoptosis of cardiac myocytes and the MTT assay was performed to determine cellular viability. Exposure to LPS significantly decreased the viability of the NRCs as well as the expression of Bcl-2, whereas it enhanced the activity and expression of the apoptosis-related proteins caspase-3 and Bax, respectively. The sarcKATP channel blocker, HMR-1098, increased the apoptosis of NRCs, whereas the specific sarcKATP channel opener, P-1075, reduced the apoptosis of NRCs. The mitochondrial calcium uniporter inhibitor ruthenium red (RR) partially inhibited the pro-apoptotic effect of HMR-1098. In order to confirm the role of the sarcKATP channel, we constructed a recombinant adenovirus vector carrying the sarcKATP channel mutant subunit Kir6.2AAA to inhibit the channel activity. Kir6.2AAA adenovirus infection in NRCs significantly aggravated the apoptosis of myocytes induced by LPS. Elucidating the regulatory mechanisms of the sarcKATP channel in apoptosis may facilitate the development of novel therapeutic targets and strategies for the management of sepsis and cardiac dysfunction.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Central Laboratory, Shenzhen Hospital Affiliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Xiaohua Zhang
- Cardiac Signaling Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Yiqun Xiong
- Central Laboratory, Shenzhen Hospital Affiliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Chaoying Xu
- Central Laboratory, Shenzhen Hospital Affiliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Xinliang Liu
- Central Laboratory, Shenzhen Hospital Affiliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Jian Lin
- Central Laboratory, Shenzhen Hospital Affiliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Guiping Mu
- Central Laboratory, Shenzhen Hospital Affiliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Shaogang Xu
- Central Laboratory, Shenzhen Hospital Affiliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Wenhe Liu
- Central Laboratory, Shenzhen Hospital Affiliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| |
Collapse
|
40
|
Liberski A, Latif N, Raynaud C, Bollensdorff C, Yacoub M. Alginate for cardiac regeneration: From seaweed to clinical trials. Glob Cardiol Sci Pract 2016; 2016:e201604. [PMID: 29043254 PMCID: PMC5642828 DOI: 10.21542/gcsp.2016.4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Heart failure is a growing endemic in the aging Western population with a prevalence of over 20 million people worldwide1. Existing heart failure therapies are unable to reverse heart failure and do not address its fundamental cause, the loss of cardiomyocytes2. In order to induce myocardial regeneration for the myocardium and the heart valve, facilitate self-repair, improve tissue salvage, reduce or reverse the adverse-remodeling and ultimately achieve long-term functional stabilization and improvement in the heart function, novel strategies for therapeutic regeneration are being developed which are aiming to compensate for the insufficient and low intrinsic regenerative ability of the adult heart3. Similarly, valve replacement with mechanical or biological substitutes meets numerous hurdles. New approaches using multicellular approaches and new material are extensively studied. Most of those strategies depend on biomaterials that help to achieve functional integrated vasculogenesis and myogenesis in the heart/tissue. Especially for failed heart valve function a number of therapeutic approaches are common from corrective intervention to complete replacement4. However the complexity of the heart valve tissue and its high physical exposure has led to a variety of approaches, however therapeutic regeneration needs to be established. Beside other approaches alginate has been identified as one building block to achieve therapeutic regeneration. Alginate is a versatile and adaptable biomaterial that has found numerous biomedical applications which include wound healing, drug delivery and tissue engineering. Due to its biologically favorable properties including the ease of gelation and its biocompatibility, alginate-based hydrogels have been considered a particularly attractive material for the application in cardiac regeneration and valve replacement techniques. Here, we review current applications of alginate in cardiac regeneration as well as perspectives for the alginate-dependent, cardiac regeneration strategies.
Collapse
Affiliation(s)
| | - Najma Latif
- Qatar Cardiovascular Research Center, Doha, Qatar
| | | | | | - Magdi Yacoub
- Qatar Cardiovascular Research Center, Doha, Qatar
| |
Collapse
|
41
|
Martins IJ. Magnesium Therapy Prevents Senescence with the Reversal of Diabetes and Alzheimer’s Disease. Health (London) 2016. [DOI: 10.4236/health.2016.87073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
42
|
Yuan Y, Zhou H, Wu QQ, Li FF, Bian ZY, Deng W, Zhou MQ, Tang QZ. Puerarin attenuates the inflammatory response and apoptosis in LPS-stimulated cardiomyocytes. Exp Ther Med 2015; 11:415-420. [PMID: 26893624 PMCID: PMC4734177 DOI: 10.3892/etm.2015.2910] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 10/26/2015] [Indexed: 01/04/2023] Open
Abstract
Patients with septic shock suffer from high mortality rates, particularly when complicated by severe myocardial depression which is characterized by hypotension and a reduction in cardiac output. Inflammation is an important factor involved in the early stages of sepsis. The aim of the present study was to investigate the effect of the Chinese herbal compound puerarin (1, 5, 10, 20 and 40 µM) on cardiomyocyte inflammatory response in a sepsis model using H9c2 cardiomyocytes stimulated with 1 µg/ml lipopolysaccharide (LPS). The mRNA expression levels of tumor necrosis factor (TNF)-α and interleukin (IL)-β were evaluated using reverse transcription-quantitative polymerase chain reaction. In addition, the protein expression levels of various factors were determined using western blot analysis. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling was used to evaluate the apoptosis rates in the various groups, and immunocytochemical analysis was employed to determine the effect of puerarin on the nuclear translocation of p65 protein. The present study demonstrated that LPS stimulation increased IL-1β and TNF-α mRNA expression levels, as compared with the controls (P<0.05). Following treatment with various concentrations of puerarin, the expression levels of IL-1β and TNF-α were markedly blunted, particularly in the LPS + 40 µM puerarin group (P<0.05 vs. the LPS group). Furthermore, puerarin administration significantly inhibited LPS-induced apoptosis in H9c2 cardiomyocytes, as determined by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining (TUNEL positive cells: LPS + 40 µM puerarin group, 5.5% vs. LPS group, 10.5%; P<0.01). In addition, puerarin significantly decreased LPS-induced phosphorylated nuclear factor (p-NF)-κB p65 and Bax expression levels, and increased the expression levels of Bcl-2, as compared with the LPS group (P<0.05). These data indicated that puerarin may serve as a valuable protective agent against cardiovascular inflammatory diseases.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Heng Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing-Qing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fang-Fang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhou-Yan Bian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Meng-Qiao Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
43
|
CYP-epoxygenase metabolites of docosahexaenoic acid protect HL-1 cardiac cells against LPS-induced cytotoxicity Through SIRT1. Cell Death Discov 2015; 1. [PMID: 27182450 PMCID: PMC4864499 DOI: 10.1038/cddiscovery.2015.54] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Bacterial LPS is an environmental toxin capable of promoting various cardiac complications. Current evidence suggests that LPS-induced myocardial dysfunction emerges as a consequence of compromised quality of cardiac mitochondria. Docosahexaenoic acid (DHA, 22:6n3) is an n-3 polyunsaturated fatty acid (PUFA), which produces a broad spectrum of intrinsic physiological effects including regulation of cell survival and death mechanisms. Although, numerous studies revealed fundamentally beneficial effects of DHA on cardiovascular system, it remains unknown whether these effects were produced by DHA or one of its possibly more potent metabolites. Emerging evidence indicates that cytochrome P450 (CYP) epoxygenase metabolites of DHA, epoxydocosapentaenoic acids (EDPs), produce more potent biological activity compared to its precursor DHA. In this study, we investigated whether DHA and its metabolite 19,20-EDP could protect HL-1 cardiac cells against LPS-induced cytotoxicity. We provide evidence that exogenously added or DHA-derived EDPs promote mitochondrial biogenesis and function in HL-1 cardiac cells. Our results illustrate the CYP epoxygenase metabolite of DHA, 19,20-EDP, confers extensive protection to HL-1 cardiac cells against LPS-induced cytotoxicity via activation of SIRT1.
Collapse
|
44
|
Trøseid M, Ueland T, Hov JR, Svardal A, Gregersen I, Dahl CP, Aakhus S, Gude E, Bjørndal B, Halvorsen B, Karlsen TH, Aukrust P, Gullestad L, Berge RK, Yndestad A. Microbiota-dependent metabolite trimethylamine-N-oxide is associated with disease severity and survival of patients with chronic heart failure. J Intern Med 2015; 277:717-26. [PMID: 25382824 DOI: 10.1111/joim.12328] [Citation(s) in RCA: 344] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Recent metabolomic, experimental and clinical studies have demonstrated that trimethylamine-N-oxide (TMAO), a microbiota-dependent metabolite from dietary phosphatidylcholine and carnitine, is a strong predictor of coronary artery disease (CAD). This finding suggests a link between the gut microbiota and atherosclerosis. The potential impact of TMAO in chronic heart failure (HF) is unknown. We hypothesized that TMAO levels would provide prognostic information about adverse outcomes in chronic HF. DESIGN Prospective, observational study including 155 consecutive patients with chronic HF. In addition, 100 patients with stable CAD without HF and 33 matched healthy individuals were included as controls. Plasma levels of TMAO and its precursors choline and betaine were measured, and associations with symptoms, aetiology and transplant-free survival in the patients with HF were explored. RESULTS Plasma levels of TMAO (P = 0.01), choline (P < 0.001) and betaine (P < 0.001) were elevated in patients with chronic HF compared to control subjects, with the highest levels in patients with New York Heart Association (NYHA) classes III and IV. Furthermore, TMAO levels were highest in individuals with ischaemic HF, followed by those with stable CAD and nonischaemic HF. TMAO, but not choline or betaine, was associated with reduced transplant-free survival: approximately 50% of patients in the upper tertile of TMAO levels died or received a heart transplant during 5.2 years of follow-up (unadjusted Cox-regression: hazard ratio 2.24, 95% confidence interval 1.28-3.92, P = 0.005). CONCLUSIONS TMAO levels were elevated in patients with HF and associated with NYHA class, ischaemic aetiology and adverse outcomes. Future studies should focus on gut microbiota, dietary composition and intestinal dysfunction in relation to TMAO levels and clinical outcome in HF.
Collapse
Affiliation(s)
- M Trøseid
- Department of Infectious Diseases, Oslo University Hospital, Ullevål, Norway.,Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway.,K.G. Jebsen Centre for Inflammation Research, Norway.,Institute of Clinical Medicine, University of Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Norway
| | - T Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway.,K.G. Jebsen Centre for Inflammation Research, Norway.,Institute of Clinical Medicine, University of Oslo, Norway
| | - J R Hov
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway.,K.G. Jebsen Centre for Inflammation Research, Norway.,Institute of Clinical Medicine, University of Oslo, Norway.,Department of Transplantation Medicine, Section of Gastroenterology, Oslo University Hospital, Norway.,Norwegian PSC Research Center, Norway
| | - A Svardal
- Department of Clinical Science, University of Bergen, Norway
| | - I Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway.,Institute of Clinical Medicine, University of Oslo, Norway
| | - C P Dahl
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway.,Department of Cardiology, Oslo University Hospital, Rikshospitalet, Norway.,Centre for Heart Failure Research, Oslo University Hospital, Norway
| | - S Aakhus
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Norway
| | - E Gude
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Norway
| | - B Bjørndal
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - B Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway.,K.G. Jebsen Centre for Inflammation Research, Norway.,Institute of Clinical Medicine, University of Oslo, Norway
| | - T H Karlsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway.,K.G. Jebsen Centre for Inflammation Research, Norway.,Institute of Clinical Medicine, University of Oslo, Norway.,Department of Transplantation Medicine, Section of Gastroenterology, Oslo University Hospital, Norway.,Norwegian PSC Research Center, Norway.,Division of Gastroenterology, Institute of Medicine, University of Bergen, Bergen, Norway
| | - P Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway.,K.G. Jebsen Centre for Inflammation Research, Norway.,Institute of Clinical Medicine, University of Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Norway
| | - L Gullestad
- Institute of Clinical Medicine, University of Oslo, Norway.,Department of Clinical Science, University of Bergen, Norway.,Centre for Heart Failure Research, Oslo University Hospital, Norway.,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway.,K.G. Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - R K Berge
- Department of Clinical Science, University of Bergen, Norway.,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - A Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway.,K.G. Jebsen Centre for Inflammation Research, Norway.,Institute of Clinical Medicine, University of Oslo, Norway.,Centre for Heart Failure Research, Oslo University Hospital, Norway
| |
Collapse
|
45
|
Oudemans-van Straaten HM, Forni LG, Groeneveld AJ, Bagshaw SM, Joannidis M. Kidney-Organ Interaction. ACUTE NEPHROLOGY FOR THE CRITICAL CARE PHYSICIAN 2015. [PMCID: PMC7123740 DOI: 10.1007/978-3-319-17389-4_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The practice of critical care nephrology demands an intimate understanding of the interactions and “crosstalk” that occurs between the kidney and multiple organ systems, in particular the heart, lung, gut, and brain. Accumulating evidence suggests that acute injury and dysfunction to the kidney can incite and propagate cardiac, pulmonary, gastrointestinal, and neurologic injury and dysfunction through a host of mechanisms.
Collapse
Affiliation(s)
| | - Lui G. Forni
- Department of Intensive Care Medicine, Anaesthesia Critical Care Collaborative, Research Group (SPACeR) and Faculty of Health Care Sciences, Royal Surrey County Hospital NHS, Foundation Trust, Surrey Perioperative, University of Surrey, Guildford, United Kingdom
| | | | - Sean M. Bagshaw
- Department of Critical Care Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta Canada
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Department of General Internal Medicine, Medical University Innsbruck, Anichstrasse, Innsbruck, Austria
| |
Collapse
|
46
|
Abstract
Cardiovascular disease is the most common cause of the greatly elevated rates of mortality characteristic of patients undergoing maintenance hemodialysis. This article is an attempt to describe the complex and evolving features of cardiac disease routinely encountered in HD patients. Furthermore, by trying to appreciate the pathophysiological drivers, and the crucial interaction with the HD treatment itself, this article seeks to define cardiac disease in this setting (HD-associated cardiomyopathy) as a unique and complex entity. By understanding the phenotype and basis of HD-associated cardiomyopathy, we can develop an evolved understanding of the dominant processes involved in its development and offer up dialysis-based interventions specifically designed to mitigate the cumulative ischemic insults consequent to conventional HD treatment. This article explores the justification of this approach and recent evidence of its efficacy.
Collapse
|
47
|
Curcumin and chronic kidney disease (CKD): major mode of action through stimulating endogenous intestinal alkaline phosphatase. Molecules 2014; 19:20139-56. [PMID: 25474287 PMCID: PMC6271001 DOI: 10.3390/molecules191220139] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 11/12/2014] [Accepted: 11/17/2014] [Indexed: 12/22/2022] Open
Abstract
Curcumin, an active ingredient in the traditional herbal remedy and dietary spice turmeric (Curcuma longa), has significant anti-inflammatory properties. Chronic kidney disease (CKD), an inflammatory disease, can lead to end stage renal disease resulting in dialysis and transplant. Furthermore, it is frequently associated with other inflammatory disease such as diabetes and cardiovascular disorders. This review will focus on the clinically relevant inflammatory molecules that play a role in CKD and associated diseases. Various enzymes, transcription factors, growth factors modulate production and action of inflammatory molecules; curcumin can blunt the generation and action of these inflammatory molecules and ameliorate CKD as well as associated inflammatory disorders. Recent studies have shown that increased intestinal permeability results in the leakage of pro-inflammatory molecules (cytokines and lipopolysaccharides) from gut into the circulation in diseases such as CKD, diabetes and atherosclerosis. This change in intestinal permeability is due to decreased expression of tight junction proteins and intestinal alkaline phosphatase (IAP). Curcumin increases the expression of IAP and tight junction proteins and corrects gut permeability. This action reduces the levels of circulatory inflammatory biomolecules. This effect of curcumin on intestine can explain why, despite poor bioavailability, curcumin has potential anti-inflammatory effects in vivo and beneficial effects on CKD.
Collapse
|
48
|
Jefferies HJ, Crowley LE, Harrison LE, Szeto CC, Li PK, Schiller B, Moran J, McIntyre CW. Circulating Endotoxaemia and Frequent Haemodialysis Schedules. ACTA ACUST UNITED AC 2014; 128:141-6. [DOI: 10.1159/000366519] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 08/04/2014] [Indexed: 11/19/2022]
|
49
|
Samokhvalov V, Vriend J, Jamieson KL, Akhnokh MK, Manne R, Falck JR, Seubert JM. PPARγ signaling is required for mediating EETs protective effects in neonatal cardiomyocytes exposed to LPS. Front Pharmacol 2014; 5:242. [PMID: 25426073 PMCID: PMC4227494 DOI: 10.3389/fphar.2014.00242] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/23/2014] [Indexed: 12/30/2022] Open
Abstract
Lipopolysaccharide (LPS) is a bacterial wall endotoxin producing many pathophysiological conditions including myocardial inflammation leading to cardiotoxicity. Epoxyeicosatrienoic acids (EETs) are biologically active metabolites of arachidonic acids capable of activating protective cellular pathways in response to stress stimuli. EETs evoke a plethora of pathways limiting impairments of cellular structures, reducing cell death, and promoting anti-inflammatory reactions in various cell types. Considering EETs are capable of producing various biological protective effects, we hypothesized that EETs would protect rat neonatal cardiomyocytes (NCM) against LPS-induced cytotoxicity. In this study, we used a dual-acting, synthetic analog of EETs, UA-8 [13-(3-propylureido)tridec-8-enoic acid], possessing both EET-mimetic and soluble epoxide hydrolase selective inhibitory properties and 14,15-EET as a model of canonical EET molecules. We found that both UA-8 and 14,15-EET significantly improved cell viability and mitochondrial function of cardiomyocytes exposed to LPS. Furthermore, treatment with UA-8 or 14,15-EET resulted in significant attenuation of LPS-triggered pro-inflammatory response, caspase-3 activation and reduction in the total antioxidant capacity in cardiomyocytes. Importantly, EET-mediated effects were significantly reduced by pharmacological inhibition of peroxisome proliferator-activated receptors γ (PPARγ) suggesting that PPARγ signaling was required for EETs exerted protective effects. Data presented in the current study demonstrate that activation of PPARγ signaling plays a crucial role in EET-mediated protection against LPS-cytotoxicity in cardiomyocytes.
Collapse
Affiliation(s)
- Victor Samokhvalov
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta Edmonton, AB, Canada
| | - Jelle Vriend
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta Edmonton, AB, Canada ; Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, VU University Amsterdam, Netherlands
| | - Kristi L Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta Edmonton, AB, Canada
| | - Maria K Akhnokh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta Edmonton, AB, Canada
| | - Rajkumar Manne
- Department of Biochemistry and Pharmacology, University of Texas Southwestern Medical Center Dallas, TX, USA
| | - John R Falck
- Department of Biochemistry and Pharmacology, University of Texas Southwestern Medical Center Dallas, TX, USA
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta Edmonton, AB, Canada ; Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta Edmonton, AB, Canada
| |
Collapse
|
50
|
Cardiopulmonary exercise capacity and preoperative markers of inflammation. Mediators Inflamm 2014; 2014:727451. [PMID: 25061264 PMCID: PMC4098894 DOI: 10.1155/2014/727451] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 12/18/2022] Open
Abstract
Explanatory mechanisms for the association between poor exercise capacity and infections following surgery are underexplored. We hypothesized that aerobic fitness—assessed by cardiopulmonary exercise testing (CPET)—would be associated with circulating inflammatory markers, as quantified by the neutrophil-lymphocyte ratio (NLR) and monocyte subsets. The association between cardiopulmonary reserve and inflammation was tested by multivariable regression analysis with covariates including anaerobic threshold (AT) and malignancy. In a first cohort of 240 colorectal patients, AT was identified as the sole factor associated with higher NLR (P = 0.03) and absolute and relative lymphopenia (P = 0.01). Preoperative leukocyte subsets and monocyte CD14+ expression (downregulated by endotoxin and indicative of chronic inflammation) were also assessed in two further cohorts of age-matched elective gastrointestinal and orthopaedic surgical patients. Monocyte CD14+ expression was lower in gastrointestinal patients (n = 43) compared to age-matched orthopaedic patients (n = 31). The circulating CD14+CD16− monocyte subset was reduced in patients with low cardiopulmonary reserve. Poor exercise capacity in patients without a diagnosis of heart failure is independently associated with markers of inflammation. These observations suggest that preoperative inflammation associated with impaired cardiorespiratory performance may contribute to the pathophysiology of postoperative outcome.
Collapse
|