1
|
Borges A, Bento L. Organ crosstalk and dysfunction in sepsis. Ann Intensive Care 2024; 14:147. [PMID: 39298039 DOI: 10.1186/s13613-024-01377-0] [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: 03/30/2024] [Accepted: 09/10/2024] [Indexed: 09/21/2024] Open
Abstract
Sepsis is a dysregulated immune response to an infection that leads to organ dysfunction. Sepsis-associated organ dysfunction involves multiple inflammatory mechanisms and complex metabolic reprogramming of cellular function. These mechanisms cooperate through multiple organs and systems according to a complex set of long-distance communications mediated by cellular pathways, solutes, and neurohormonal actions. In sepsis, the concept of organ crosstalk involves the dysregulation of one system, which triggers compensatory mechanisms in other systems that can induce further damage. Despite the abundance of studies published on organ crosstalk in the last decade, there is a need to formulate a more comprehensive framework involving all organs to create a more detailed picture of sepsis. In this paper, we review the literature published on organ crosstalk in the last 10 years and explore how these relationships affect the progression of organ failure in patients with septic shock. We explored these relationships in terms of the heart-kidney-lung, gut-microbiome-liver-brain, and adipose tissue-muscle-bone crosstalk in sepsis patients. A deep connection exists among these organs based on crosstalk. We also review how multiple therapeutic interventions administered in intensive care units, such as mechanical ventilation, antibiotics, anesthesia, nutrition, and proton pump inhibitors, affect these systems and must be carefully considered when managing septic patients. The progression to multiple organ dysfunction syndrome in sepsis patients is still one of the most frequent causes of death in critically ill patients. A better understanding and monitoring of the mechanics of organ crosstalk will enable the anticipation of organ damage and the development of individualized therapeutic strategies.
Collapse
Affiliation(s)
- André Borges
- Intensive Care Unit of Hospital de São José, Unidade de Urgência Médica, Rua José António Serrano, Lisbon, 1150-199, Portugal.
- NOVA Medical School, Campo dos Mártires da Pátria 130, Lisbon, 1169-056, Portugal.
| | - Luís Bento
- Intensive Care Unit of Hospital de São José, Unidade de Urgência Médica, Rua José António Serrano, Lisbon, 1150-199, Portugal
- NOVA Medical School, Campo dos Mártires da Pátria 130, Lisbon, 1169-056, Portugal
| |
Collapse
|
2
|
Lu KC, Tsai KW, Hu WC. Role of TGFβ-producing regulatory T cells in scleroderma and end-stage organ failure. Heliyon 2024; 10:e35590. [PMID: 39170360 PMCID: PMC11336735 DOI: 10.1016/j.heliyon.2024.e35590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
Regulatory T cells (Tregs) are crucial immune cells that initiate a tolerable immune response. Transforming growth factor-beta (TGFβ) is a key cytokine produced by Tregs and plays a significant role in stimulating tissue fibrosis. Systemic sclerosis, an autoimmune disease characterized by organ fibrosis, is associated with an overrepresentation of regulatory T cells. This review aims to identify Treg-dominant tolerable host immune reactions and discuss their association with scleroderma and end-stage organ failure. End-stage organ failures, including heart failure, liver cirrhosis, uremia, and pulmonary fibrosis, are frequently linked to tissue fibrosis. This suggests that TGFβ-producing Tregs are involved in the pathogenesis of these conditions. However, the exact significance of TGFβ and the mechanisms through which it induces tolerable immune reactions during end-stage organ failure remain unclear. A deeper understanding of these mechanisms could lead to improved preventive and therapeutic strategies for these severe diseases.
Collapse
Affiliation(s)
- Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Kuo-Wang Tsai
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, 231, Taiwan
| | - Wan-Chung Hu
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, 231, Taiwan
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, 231, Taiwan
- Department of Biotechnology, Ming Chuan University, Taoyuan City, 333, Taiwan
| |
Collapse
|
3
|
Zandawala M, Gera J. Leptin- and cytokine-like unpaired signaling in Drosophila. Mol Cell Endocrinol 2024; 584:112165. [PMID: 38266772 DOI: 10.1016/j.mce.2024.112165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
Abstract
Animals have evolved a multitude of signaling pathways that enable them to orchestrate diverse physiological processes to tightly regulate systemic homeostasis. This signaling is mediated by various families of peptide hormones and cytokines that are conserved across the animal kingdom. In this review, we primarily focus on the unpaired (Upd) family of proteins in Drosophila which are evolutionarily related to mammalian leptin and the cytokine interleukin 6. We summarize expression patterns of Upd in Drosophila and discuss the parallels in structure, signaling pathway, and functions between Upd and their mammalian counterparts. In particular, we focus on the roles of Upd in governing metabolic homeostasis, growth and development, and immune responses. We aim to stimulate future studies on leptin-like signaling in other phyla which can help bridge the evolutionary gap between insect Upd and vertebrate leptin and cytokines like interleukin 6.
Collapse
Affiliation(s)
- Meet Zandawala
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany; Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, 89557, USA.
| | - Jayati Gera
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| |
Collapse
|
4
|
Dinh H, Kovács ZZA, Kis M, Kupecz K, Sejben A, Szűcs G, Márványkövi F, Siska A, Freiwan M, Pósa SP, Galla Z, Ibos KE, Bodnár É, Lauber GY, Goncalves AIA, Acar E, Kriston A, Kovács F, Horváth P, Bozsó Z, Tóth G, Földesi I, Monostori P, Cserni G, Podesser BK, Lehoczki A, Pokreisz P, Kiss A, Dux L, Csabafi K, Sárközy M. Role of the kisspeptin-KISS1R axis in the pathogenesis of chronic kidney disease and uremic cardiomyopathy. GeroScience 2024; 46:2463-2488. [PMID: 37987885 PMCID: PMC10828495 DOI: 10.1007/s11357-023-01017-8] [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: 06/13/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
The prevalence of chronic kidney disease (CKD) is increasing globally, especially in elderly patients. Uremic cardiomyopathy is a common cardiovascular complication of CKD, characterized by left ventricular hypertrophy (LVH), diastolic dysfunction, and fibrosis. Kisspeptins and their receptor, KISS1R, exert a pivotal influence on kidney pathophysiology and modulate age-related pathologies across various organ systems. KISS1R agonists, including kisspeptin-13 (KP-13), hold promise as novel therapeutic agents within age-related biological processes and kidney-related disorders. Our investigation aimed to elucidate the impact of KP-13 on the trajectory of CKD and uremic cardiomyopathy. Male Wistar rats (300-350 g) were randomized into four groups: (I) sham-operated, (II) 5/6 nephrectomy-induced CKD, (III) CKD subjected to a low dose of KP-13 (intraperitoneal 13 µg/day), and (IV) CKD treated with a higher KP-13 dose (intraperitoneal 26 µg/day). Treatments were administered daily from week 3 for 10 days. After 13 weeks, KP-13 increased systemic blood pressure, accentuating diastolic dysfunction's echocardiographic indicators and intensifying CKD-associated markers such as serum urea levels, glomerular hypertrophy, and tubular dilation. Notably, KP-13 did not exacerbate circulatory uremic toxin levels, renal inflammation, or fibrosis markers. In contrast, the higher KP-13 dose correlated with reduced posterior and anterior wall thickness, coupled with diminished cardiomyocyte cross-sectional areas and concurrent elevation of inflammatory (Il6, Tnf), fibrosis (Col1), and apoptosis markers (Bax/Bcl2) relative to the CKD group. In summary, KP-13's influence on CKD and uremic cardiomyopathy encompassed heightened blood pressure and potentially activated inflammatory and apoptotic pathways in the left ventricle.
Collapse
Affiliation(s)
- Hoa Dinh
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
- Department of Biochemistry, Bach Mai Hospital, Hanoi, 100000, Vietnam
| | - Zsuzsanna Z A Kovács
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Merse Kis
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Klaudia Kupecz
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Anita Sejben
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Gergő Szűcs
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Fanni Márványkövi
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Andrea Siska
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Marah Freiwan
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Szonja Polett Pósa
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Zsolt Galla
- Metabolic and Newborn Screening Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Katalin Eszter Ibos
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Éva Bodnár
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Gülsüm Yilmaz Lauber
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Ana Isabel Antunes Goncalves
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Eylem Acar
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - András Kriston
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, 6726, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, 6726, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Ferenc Kovács
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, 6726, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, 6726, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Péter Horváth
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, 6726, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, 6726, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Zsolt Bozsó
- Department of Medical Chemistry, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Gábor Tóth
- Department of Medical Chemistry, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Péter Monostori
- Metabolic and Newborn Screening Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Andrea Lehoczki
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
| | - Peter Pokreisz
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - László Dux
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary.
| | - Krisztina Csabafi
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Márta Sárközy
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary.
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary.
| |
Collapse
|
5
|
Tazarghi A, Bazoq S, Taziki Balajelini MH, Ebrahimi M, Hosseini SM, Razavi Nikoo H. Liver injury in COVID-19: an insight into pathobiology and roles of risk factors. Virol J 2024; 21:65. [PMID: 38491495 PMCID: PMC10943793 DOI: 10.1186/s12985-024-02332-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/27/2024] [Indexed: 03/18/2024] Open
Abstract
COVID-19 is a complex disease that can lead to fatal respiratory failure with extrapulmonary complications, either as a direct result of viral invasion in multiple organs or secondary to oxygen supply shortage. Liver is susceptible to many viral pathogens, and due to its versatile functions in the body, it is of great interest to determine how hepatocytes may interact with SARS-CoV-2 in COVID-19 patients. Liver injury is a major cause of death, and SARS-CoV-2 is suspected to contribute significantly to hepatopathy. Owing to the lack of knowledge in this field, further research is required to address these ambiguities. Therefore, we aimed to provide a comprehensive insight into host-virus interactions, underlying mechanisms, and associated risk factors by collecting results from epidemiological analyses and relevant laboratory experiments. Backed by an avalanche of recent studies, our findings support that liver injury is a sequela of severe COVID-19, and certain pre-existing liver conditions can also intensify the morbidity of SARS-CoV-2 infection in synergy. Notably, age, sex, lifestyle, dietary habits, coinfection, and particular drug regimens play a decisive role in the final outcome and prognosis as well. Taken together, our goal was to unravel these complexities concerning the development of novel diagnostic, prophylactic, and therapeutic approaches with a focus on prioritizing high-risk groups.
Collapse
Affiliation(s)
- Abbas Tazarghi
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sahar Bazoq
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohammad Hosein Taziki Balajelini
- Department of Otorhinolaryngology, Neuroscience Research Center, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohsen Ebrahimi
- Neonatal and Children's Health Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Seyed Mehran Hosseini
- Department of Physiology, School of Medicine, Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Hadi Razavi Nikoo
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
6
|
Walther CP, Civitello AB, Lamba HK, Mondal NK, Navaneethan SD. Kidney Function Trajectories and Right Heart Failure Following LVAD Implantation. J Am Heart Assoc 2024; 13:e031305. [PMID: 38420763 PMCID: PMC10944080 DOI: 10.1161/jaha.123.031305] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 12/01/2023] [Indexed: 03/02/2024]
Abstract
BACKGROUND Preoperative kidney dysfunction is a risk factor for right heart failure (RHF) after implantation of a left ventricular assist device (LVAD). However, characteristic kidney function trajectories before and after post-LVAD RHF are uncertain, so we investigated this. METHODS AND RESULTS We identified individuals who received primary continuous-flow LVAD implantation from July 1, 2014 to December 31, 2017 in the Interagency Registry for Mechanically Assisted Circulatory Support (INTERMACS) data set. Incident RHF was ascertained using the INTERMACS definition at 1 and 3 months and classified as transient or persistent. Kidney function trajectories before and after RHF onset, and relationships of baseline kidney function with RHF risk at the different time points, were assessed. We identified 8076 LVAD recipients who met inclusion criteria. Incident RHF was present at 1 month in 26.4%. There were 4850 individuals with follow-up at 3 months, with incident RHF in 4.2%. Kidney function trajectories differed from pre-LVAD implantation to 1-month follow-up by RHF category, with those developing persistent RHF having no improvement in baseline kidney function. For trajectories before the 3-month RHF ascertainment time, the shape was similar for those with and without RHF, with lower estimated glomerular filtration rate levels among those who developed RHF. Baseline estimated glomerular filtration rate levels below the normal range were associated with higher risk of RHF at 1 and 3 months. CONCLUSIONS In LVAD recipients, preimplantation kidney function and subsequent kidney function trajectories differed substantially by RHF at 1 and 3 months postimplantation, even after adjustment for several confounders. This may demonstrate bidirectional associations between kidney function and right ventricular function in LVAD recipients.
Collapse
Affiliation(s)
- Carl P. Walther
- Selzman Institute for Kidney Health, Section of Nephrology, Department of MedicineBaylor College of MedicineHoustonTX
| | - Andrew B. Civitello
- Section of Cardiology, Department of MedicineBaylor College of MedicineHoustonTX
- Advanced Heart Failure Center of ExcellenceBaylor College of MedicineHoustonTX
| | - Harveen K. Lamba
- Division of Cardiothoracic Transplantation and Circulatory Support, Department of SurgeryBaylor College of MedicineHoustonTX
| | - Nandan K. Mondal
- Division of Cardiothoracic Transplantation and Circulatory Support, Department of SurgeryBaylor College of MedicineHoustonTX
| | - Sankar D. Navaneethan
- Selzman Institute for Kidney Health, Section of Nephrology, Department of MedicineBaylor College of MedicineHoustonTX
- Section of NephrologyMichael E. DeBakey Veterans Affairs Medical CenterHoustonTX
- Institute of Clinical and Translational Research, Baylor College of MedicineHoustonTX
| |
Collapse
|
7
|
Joseph A, Petit M, Vieillard-Baron A. Hemodynamic effects of positive end-expiratory pressure. Curr Opin Crit Care 2024; 30:10-19. [PMID: 38085886 DOI: 10.1097/mcc.0000000000001124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
PURPOSE OF REVIEW Positive end-expiratory pressure (PEEP) is required in the Berlin definition of acute respiratory distress syndrome and is a cornerstone of its treatment. Application of PEEP increases airway pressure and modifies pleural and transpulmonary pressures according to respiratory mechanics, resulting in blood volume alteration into the pulmonary circulation. This can in turn affect right ventricular preload, afterload and function. At the opposite, PEEP may improve left ventricular function, providing no deleterious effect occurs on the right ventricle. RECENT FINDINGS This review examines the impact of PEEP on cardiac function with regards to heart-lung interactions, and describes its consequences on organs perfusion and function, including the kidney, gut, liver and the brain. PEEP in itself is not beneficious nor detrimental on end-organ hemodynamics, but its hemodynamic effects vary according to both respiratory mechanics and association with other hemodynamic variables such as central venous or mean arterial pressure. There are parallels in the means of preventing deleterious impact of PEEP on the lungs, heart, kidney, liver and central nervous system. SUMMARY The quest for optimal PEEP settings has been a prominent goal in ARDS research for the last decades. Intensive care physician must maintain a high degree of vigilance towards hemodynamic effects of PEEP on cardiac function and end-organs circulation.
Collapse
Affiliation(s)
- Adrien Joseph
- Medical Intensive Care Unit, Ambroise Paré Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt
| | - Matthieu Petit
- Medical Intensive Care Unit, Ambroise Paré Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt
- Inserm, CESP, Paris-Saclay University, Université de Versailles Saint-Quentin-en-Yvelines, Villejuif, France
| | - Antoine Vieillard-Baron
- Medical Intensive Care Unit, Ambroise Paré Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt
- Inserm, CESP, Paris-Saclay University, Université de Versailles Saint-Quentin-en-Yvelines, Villejuif, France
| |
Collapse
|
8
|
Battaglini D, De Rosa S, Godoy DA. Crosstalk Between the Nervous System and Systemic Organs in Acute Brain Injury. Neurocrit Care 2024; 40:337-348. [PMID: 37081275 DOI: 10.1007/s12028-023-01725-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 03/29/2023] [Indexed: 04/22/2023]
Abstract
Organ crosstalk is a complex biological communication between distal organs mediated via cellular, soluble, and neurohormonal actions, based on a two-way pathway. The communication between the central nervous system and peripheral organs involves nerves, endocrine, and immunity systems as well as the emotional and cognitive centers of the brain. Particularly, acute brain injury is complicated by neuroinflammation and neurodegeneration causing multiorgan inflammation, microbial dysbiosis, gastrointestinal dysfunction and dysmotility, liver dysfunction, acute kidney injury, and cardiac dysfunction. Organ crosstalk has become increasingly popular, although the information is still limited. The present narrative review provides an update on the crosstalk between the nervous system and systemic organs after acute brain injury. Future research might help to target this pathophysiological process, preventing the progression toward multiorgan dysfunction in critically ill patients with brain injury.
Collapse
Affiliation(s)
- Denise Battaglini
- Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia De Rosa
- Centre for Medical Sciences, University of Trento, Via S. Maria Maddalena 1, 38122, Trento, Italy.
- Anesthesia and Intensive Care, Santa Chiara Regional Hospital, APSS Trento, Trento, Italy.
| | | |
Collapse
|
9
|
Endo A, Hirose T, Sato S, Ito H, Takahashi C, Ishikawa R, Kamada A, Oba-Yabana I, Kimura T, Takahashi K, Mori T. Sodium glucose cotransporter 2 inhibitor suppresses renal injury in rats with renal congestion. Hypertens Res 2024; 47:33-45. [PMID: 37749334 PMCID: PMC10766540 DOI: 10.1038/s41440-023-01437-1] [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: 07/31/2023] [Revised: 09/10/2023] [Accepted: 09/10/2023] [Indexed: 09/27/2023]
Abstract
Renal congestion is an issue of cardiorenal syndrome in patients with heart failure. Recent clinical and basic studies suggest a renoprotective potential of sodium-glucose cotransporter (SGLT) 2 inhibitors. However, the effect on renal congestion and its mechanism is not fully understood. Thus, we aimed to clarify the effect of SGLT inhibition in a renal congestion model. Renal congestion was induced in the left kidney of male Sprague-Dawley rats by ligation of the inferior vena cava between the renal veins. The SGLT2 inhibitor tofogliflozin or vehicle was orally administered daily from the day before IVC ligation until two days after surgery. On the third postoperative day, both the right control kidney and the left congested kidney were harvested and analyzed. Kidney weight and water content was increased, and renal injury and fibrosis were observed in the left congested kidney. Kidney weight gain and hydration were improved with tofogliflozin treatment. Additionally, this treatment effectively reduced renal injury and fibrosis, particularly in the renal cortex. SGLT2 expression was observed in the congested kidney, but suppressed in the damaged tubular cells. Molecules associated with inflammation were increased in the congested kidney and reversed by tofogliflozin treatment. Mitochondrial dysfunction provoked by renal congestion was also improved by tofogliflozin treatment. Tofogliflozin protects against renal damage induced by renal congestion. SGLT2 inhibitors could be a candidate strategy for renal impairment associated with heart failure.
Collapse
Affiliation(s)
- Akari Endo
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takuo Hirose
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan.
- Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
| | - Shigemitsu Sato
- Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Hiroki Ito
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chika Takahashi
- Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Risa Ishikawa
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ayaka Kamada
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ikuko Oba-Yabana
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tomoyoshi Kimura
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kazuhiro Takahashi
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takefumi Mori
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
- Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
| |
Collapse
|
10
|
Karumanchi P, Sridharan D, Hoppensteadt D, Siddiqui F, Fareed J, Bansal V. Thromboinflammatory Biomarkers of Cardiorenal Syndrome in Patients With End-Stage Renal Disease. Clin Appl Thromb Hemost 2024; 30:10760296241263101. [PMID: 38863224 PMCID: PMC11179552 DOI: 10.1177/10760296241263101] [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: 03/14/2024] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
Cardiovascular disease is a prevalent complication in patients with end-stage renal disease (ESRD) on maintenance hemodialysis. In the ESRD patient population, cardiovascular mortality is 20 times higher compared to the general population. The strong relationship between both illnesses can be explained through cardiorenal syndrome (CRS). CRS encompasses a spectrum of disorders involving both the heart and kidneys in which acute or chronic dysfunction in one organ may induce a similar effect in the other organ. Current literature reveals that inflammation and thrombosis are integral to CRS development. Hence, this study aims to demonstrate whether thromboinflammatory biomarkers and laboratory parameters correlate with ESRD progression and the development of CRS. Ninety-five ESRD patients were recruited at Loyola University Medical Center hemodialysis unit. Epic chart analysis was used to determine patients with CRS. Biomarkers (C-reactive protein, tumor necrosis factor alpha, interleukin-6, Annexin V, L-fatty acid binding protein, monocyte chemoattractant protein 1, nitric oxide, von Willebrand factor, D-dimer, and plasminogen activator inhibitor-1) were profiled using the enzyme-linked immunosorbent assay method in patients with and without CRS in the ESRD cohort. All biomarkers were significantly elevated in ESRD patients compared to normal controls (P < .05) and laboratory parameters, ferritin (521.99 ± 289.33) and PTH (442.91 ± 1.50). Through EPIC chart analysis 47% of ESRD patients have CRS. D-dimer and TNF-α were significantly elevated in patients with CRS compared to patients without CRS. This study suggests that biomarkers, D-dimer, and TNF-α, can be good predictors of CRS in ESRD patients.
Collapse
Affiliation(s)
| | - Divya Sridharan
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Debra Hoppensteadt
- Department of Pathology & Laboratory Medicine, and Department of Pharmacology and Neuroscience, Cardiovascular Research Institute, Health Science Division, Loyola University Chicago, Maywood, IL, USA
| | - Fakiha Siddiqui
- Program in Health Sciences, UCAM - Universidad Católica San Antonio de Murcia, Murcia, Spain
- Department of Pathology & Laboratory Medicine, Cardiovascular Research Institute, Health Science Division, Loyola University Chicago, Maywood, IL, USA
| | - Jawed Fareed
- Department of Pathology & Laboratory Medicine, and Department of Pharmacology and Neuroscience, Cardiovascular Research Institute, Health Science Division, Loyola University Chicago, Maywood, IL, USA
| | - Vinod Bansal
- Department of Nephrology, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
11
|
Cepoi MR, Duca ST, Chetran A, Costache AD, Spiridon MR, Afrăsânie I, Leancă SA, Dmour BA, Matei IT, Miftode RS, Miftode L, Prepeliuc CS, Haba MȘC, Bădescu MC, Costache II. Chronic Kidney Disease Associated with Ischemic Heart Disease: To What Extent Do Biomarkers Help? Life (Basel) 2023; 14:34. [PMID: 38255650 PMCID: PMC10817293 DOI: 10.3390/life14010034] [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: 11/20/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Chronic kidney disease represents a complex and multifaceted pathology characterized by the presence of structural or functional renal anomalies associated with a persistent reduction in renal function. As the disease progresses, complications arise due to the chronic inflammatory syndrome, hydro-electrolytic disorders, and toxicity secondary to the uremic environment. Cardiovascular complications are the leading cause of death for these patients. Ischemic cardiac pathology can be both a consequence and complication of chronic kidney disease, highlighting the need to identify specific cardiorenal dysfunction biomarkers targeting pathophysiological mechanisms common to both conditions. This identification is crucial for establishing accurate diagnoses, prognoses, and risk stratifications for patients. This work is intended to elucidate the intricate relationship between chronic kidney disease and ischemic heart disease and to investigate the roles of cardiorenal biomarkers, including cardiac troponin, natriuretic peptides, galectin-3, copeptin, fibroblast growth factor 23 and its co-receptor Klotho, soluble suppression of tumorigenicity 2, and plasma growth differentiation factor 15.
Collapse
Affiliation(s)
- Maria-Ruxandra Cepoi
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Stefania Teodora Duca
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Adriana Chetran
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Alexandru Dan Costache
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Marilena Renata Spiridon
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Irina Afrăsânie
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Sabina Andreea Leancă
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Bianca-Ana Dmour
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of III Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Iulian Theodor Matei
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Radu Stefan Miftode
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Larisa Miftode
- Department of Infectious Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (L.M.); (C.S.P.)
- “St. Parascheva” Clinical Hospital of Infectious Diseases, 700116 Iași, Romania
| | - Cristian Sorin Prepeliuc
- Department of Infectious Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (L.M.); (C.S.P.)
- “St. Parascheva” Clinical Hospital of Infectious Diseases, 700116 Iași, Romania
| | - Mihai Ștefan Cristian Haba
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Minerva Codruța Bădescu
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of III Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Irina Iuliana Costache
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| |
Collapse
|
12
|
Sárközy M, Watzinger S, Kovács ZZ, Acar E, Márványkövi F, Szűcs G, Lauber GY, Galla Z, Siska A, Földesi I, Fintha A, Kriston A, Kovács F, Horváth P, Kővári B, Cserni G, Krenács T, Szabó PL, Szabó GT, Monostori P, Zins K, Abraham D, Csont T, Pokreisz P, Podesser BK, Kiss A. Neuregulin-1β Improves Uremic Cardiomyopathy and Renal Dysfunction in Rats. JACC Basic Transl Sci 2023; 8:1160-1176. [PMID: 37791301 PMCID: PMC10543921 DOI: 10.1016/j.jacbts.2023.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/01/2023] [Accepted: 03/01/2023] [Indexed: 10/05/2023]
Abstract
Chronic kidney disease is a global health problem affecting 10% to 12% of the population. Uremic cardiomyopathy is often characterized by left ventricular hypertrophy, fibrosis, and diastolic dysfunction. Dysregulation of neuregulin-1β signaling in the heart is a known contributor to heart failure. The systemically administered recombinant human neuregulin-1β for 10 days in our 5/6 nephrectomy-induced model of chronic kidney disease alleviated the progression of uremic cardiomyopathy and kidney dysfunction in type 4 cardiorenal syndrome. The currently presented positive preclinical data warrant clinical studies to confirm the beneficial effects of recombinant human neuregulin-1β in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Márta Sárközy
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Simon Watzinger
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Zsuzsanna Z.A. Kovács
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Eylem Acar
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Fanni Márványkövi
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Gergő Szűcs
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Gülsüm Yilmaz Lauber
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Zsolt Galla
- Metabolic and Newborn Screening Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, Hungary
| | - Andrea Siska
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Attila Fintha
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - András Kriston
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Ferenc Kovács
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Péter Horváth
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Bence Kővári
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tibor Krenács
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Petra Lujza Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Gábor Tamás Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Péter Monostori
- Metabolic and Newborn Screening Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, Hungary
| | - Karin Zins
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Abraham
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Tamás Csont
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Peter Pokreisz
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Bruno K. Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Basak T, Saraswati S. Editorial: Fibrotic tissue remodeling as a driver of disease pathogenesis. Front Mol Biosci 2023; 10:1278388. [PMID: 37711391 PMCID: PMC10497952 DOI: 10.3389/fmolb.2023.1278388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023] Open
Affiliation(s)
- Trayambak Basak
- School of Biosciences and Bioengineering, Indian Institute Of Technology–Mandi (IIT–Mandi), Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute Of Technology–Mandi (IIT–Mandi), Mandi, Himachal Pradesh, India
| | - Sarika Saraswati
- Department of Biological Sciences, Tennessee State University, Nashville, TN, United States
| |
Collapse
|
14
|
Dinh H, Kovács ZZA, Márványkövi F, Kis M, Kupecz K, Szűcs G, Freiwan M, Lauber GY, Acar E, Siska A, Ibos KE, Bodnár É, Kriston A, Kovács F, Horváth P, Földesi I, Cserni G, Podesser BK, Pokreisz P, Kiss A, Dux L, Csabafi K, Sárközy M. The kisspeptin-1 receptor antagonist peptide-234 aggravates uremic cardiomyopathy in a rat model. Sci Rep 2023; 13:14046. [PMID: 37640761 PMCID: PMC10462750 DOI: 10.1038/s41598-023-41037-0] [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: 01/26/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
Uremic cardiomyopathy is characterized by diastolic dysfunction, left ventricular hypertrophy (LVH), and fibrosis. Dysregulation of the kisspeptin receptor (KISS1R)-mediated pathways are associated with the development of fibrosis in cancerous diseases. Here, we investigated the effects of the KISS1R antagonist peptide-234 (P234) on the development of uremic cardiomyopathy. Male Wistar rats (300-350 g) were randomized into four groups: (i) Sham, (ii) chronic kidney disease (CKD) induced by 5/6 nephrectomy, (iii) CKD treated with a lower dose of P234 (ip. 13 µg/day), (iv) CKD treated with a higher dose of P234 (ip. 26 µg/day). Treatments were administered daily from week 3 for 10 days. At week 13, the P234 administration did not influence the creatinine clearance and urinary protein excretion. However, the higher dose of P234 led to reduced anterior and posterior wall thicknesses, more severe interstitial fibrosis, and overexpression of genes associated with left ventricular remodeling (Ctgf, Tgfb, Col3a1, Mmp9), stretch (Nppa), and apoptosis (Bax, Bcl2, Casp7) compared to the CKD group. In contrast, no significant differences were found in the expressions of apoptosis-associated proteins between the groups. Our results suggest that the higher dose of P234 hastens the development and pathophysiology of uremic cardiomyopathy by activating the fibrotic TGF-β-mediated pathways.
Collapse
Affiliation(s)
- Hoa Dinh
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
- Department of Biochemistry, Bach Mai Hospital, Hanoi, 100000, Vietnam
| | - Zsuzsanna Z A Kovács
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Fanni Márványkövi
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Merse Kis
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Klaudia Kupecz
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Gergő Szűcs
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Marah Freiwan
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Gülsüm Yilmaz Lauber
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, A1090, Vienna, Austria
| | - Eylem Acar
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, A1090, Vienna, Austria
| | - Andrea Siska
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Katalin Eszter Ibos
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Éva Bodnár
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - András Kriston
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, 6726, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, 6726, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Ferenc Kovács
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, 6726, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, 6726, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Péter Horváth
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, 6726, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, 6726, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, A1090, Vienna, Austria
| | - Peter Pokreisz
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, A1090, Vienna, Austria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, A1090, Vienna, Austria
| | - László Dux
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary.
| | - Krisztina Csabafi
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Márta Sárközy
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary.
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary.
| |
Collapse
|
15
|
Gao H, Bai M, Chu A, Pan C, Zhao J, Zhang Z, Shang X. Safety and efficacy of perioperative continuous renal replacement therapy for percutaneous coronary intervention in severe acute myocardial infarction patients. J Med Life 2023; 16:719-724. [PMID: 37520492 PMCID: PMC10375348 DOI: 10.25122/jml-2022-0270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 03/27/2023] [Indexed: 08/01/2023] Open
Abstract
This retrospective study aimed to evaluate the safety and efficacy of continuous renal replacement therapy (CRRT) during percutaneous coronary intervention (PCI) in patients with severe acute myocardial infarction (AMI). The study analyzed data from 945 AMI patients hospitalized between January 2016 and December 2017, out of which 21 patients underwent perioperative CRRT for PCI. We assessed the baseline characteristics of severe AMI patients before and after CRRT and examined the effect of CRRT on cardiac, renal, and liver function, as well as other indicators. The heart rate of patients undergoing CRRT was significantly lower at 24 h and 48 h after CRRT than before CRRT (p=0.038). There was a moderate but not significant decrease in the mean systolic blood pressure or diastolic blood pressure (p>0.05). Importantly, we found that significantly more patients showed Killip class I-II and significantly improved cardiac function after CRRT (23.8% vs. 57.1%, p=0.001). The levels of urea nitrogen, creatinine, aspartate aminotransferase, glutamic pyruvic transaminase, and total bilirubin were significantly lowered after CRRT treatment (p<0.05). Perioperative management of CRRT was safe and effective for severe AMI patients.
Collapse
Affiliation(s)
- Hanxiang Gao
- Department of Cardiology, First Hospital of Lanzhou University, Lanzhou, China
| | - Ming Bai
- Department of Cardiology, First Hospital of Lanzhou University, Lanzhou, China
| | - Aiai Chu
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, China
| | - Chenliang Pan
- Department of Cardiology, First Hospital of Lanzhou University, Lanzhou, China
| | - Jing Zhao
- Department of Cardiology, First Hospital of Lanzhou University, Lanzhou, China
| | - Zheng Zhang
- Department of Cardiology, First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaofeng Shang
- Department of Cardiology, Zhangye People's Hospital Affiliated Hexi College, Zhangye, Gansu, China
| |
Collapse
|
16
|
Zhang F, Wang H, Bai Y, Huang L, Zhang H. Effect of respiratory muscle training in patients with chronic kidney disease: A systematic review and meta-analysis of randomized controlled trials. Clin Rehabil 2023; 37:348-361. [PMID: 36325749 DOI: 10.1177/02692155221135729] [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: 11/06/2022]
Abstract
OBJECTIVE This systematic review and meta-analysis aimed to evaluate the effect of respiratory muscle training on respiratory muscle strength, lung function, cardiopulmonary fitness, and quality of life for chronic kidney disease patients. METHODS PubMed, Embase, Web of Science Core Collection, and Cochrane Central Register of Controlled Trials published randomized controlled trials that evaluated the effect of respiratory muscle training for chronic kidney disease patients from inception to December 2021, and rerun on September 2022. The quality of included studies was evaluated according to the Cochrane Collaboration's risk of bias tool-2. The outcomes were analyzed as mean differences with a fixed/random effect model. The strength of evidence was evaluated with the Grading of Recommendation, Assessment, Development, and Evaluation approach. RESULTS Eleven randomized controlled trials were included. All but two of the studies were in hemodialysis patients. The follow-up time range was 4 to 12 weeks. Compared to controls, respiratory muscle training significantly improved maximal expiratory pressure (mean difference = 17.36, p = 0.013), maximal inspiratory pressure (mean difference = 18.26, p = 0.002), forced expiratory volume at 1 second (mean difference = 0.20, p= 0.020), forced vital capacity (mean difference = 0.26, p = 0.008), but not for 6-minute walk test (mean difference = 39.85, p= 0.138). CONCLUSIONS As a non-pharmacological therapy, respiratory muscle training can effectively improve maximal expiratory pressure, maximal inspiratory pressure, forced expiratory volume at 1 second, and forced vital capacity in patients with chronic kidney disease and is safe for such populations.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Nephrology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Wang
- Department of Anorectology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Bai
- Department of Cardiology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liuyan Huang
- Department of Nephrology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huachun Zhang
- Department of Nursing, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Fernandes J, Costa R, Guerreiro R, Bonifácio D, Rodrigues A, Henriques C, Branco P, Araújo I, Fonseca C. Co-Administration of Albumin and Furosemide in Acute Heart Failure with Diuretics Resistance. ACTA MEDICA PORT 2023; 36:193-201. [PMID: 36762993 DOI: 10.20344/amp.17714] [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: 12/14/2021] [Accepted: 10/11/2022] [Indexed: 02/11/2023]
Abstract
Acute heart failure is a frequent cause of hospital admission in Portugal, and has an increasing tendency given the aging population. Although most admissions for acute heart failure are caused by congestive conditions, not all patients have a congestive phenotype, reflecting the complexity of a process with multiple pathophysiological pathways. The use of diuretics, usually loop diuretics, is the mainstay of treatment for congestion. However, many patients develop resistance, thus constituting a challenge with no consensual solution to date, despite extensive debate over the years. Despite its frequent use in clinical practice, the co-administration of albumin and furosemide remains controversial in the management of patients with acute heart failure, hypoalbuminemia, and diuretic resistance. This review addresses the pathophysiological mechanisms of congestion in patients with acute heart failure and explores the theoretical basis that supports the co-administration of albumin and furosemide in this clinical context. It is intended to clarify the potential benefit of the combined approach in this specific population and identify possible gaps in the literature that could be the subject of future studies.
Collapse
Affiliation(s)
- Jorge Fernandes
- Unidade Funcional de Medicina Interna 7.2. Hospital Curry Cabral. Centro Hospitalar Universitário de Lisboa Central. Lisboa. Portugal
| | - Rita Costa
- Serviço de Medicina Interna. Centro Hospitalar de Vila Nova de Gaia/Espinho. Vila Nova de Gaia..
| | - Renato Guerreiro
- Serviço de Medicina Interna. Hospital de São Francisco Xavier. Centro Hospitalar de Lisboa Ocidental. Lisboa. Portugal
| | - Dulce Bonifácio
- Serviço de Medicina Interna. Hospital Distrital de Torres Vedras. Centro Hospitalar do Oeste. Torres Vedras. Portugal
| | - Ana Rodrigues
- Serviço de Medicina Interna. Unidade Local de Saúde do Norte Alentejano. Hospital Santa Luzia de Elvas. Elvas. Portugal
| | - Célia Henriques
- Clínica de Insuficiência Cardíaca. Serviço de Medicina III. Hospital de São Francisco Xavier. Centro Hospitalar de Lisboa Ocidental. Lisboa; NOVA Medical School. Faculdade de Ciências Médicas. Universidade Nova de Lisboa. Lisboa. Portugal
| | - Patrícia Branco
- NOVA Medical School. Faculdade de Ciências Médicas. Universidade Nova de Lisboa. Lisboa; Serviço de Nefrologia. Hospital de Santa Cruz. Centro Hospitalar de Lisboa Ocidental. Lisboa. Portugalm
| | - Inês Araújo
- Clínica de Insuficiência Cardíaca. Serviço de Medicina III. Hospital de São Francisco Xavier. Centro Hospitalar de Lisboa Ocidental. Lisboa; NOVA Medical School. Faculdade de Ciências Médicas. Universidade Nova de Lisboa. Lisboa. Portugal
| | - Cândida Fonseca
- Clínica de Insuficiência Cardíaca. Serviço de Medicina III. Hospital de São Francisco Xavier. Centro Hospitalar de Lisboa Ocidental. Lisboa; NOVA Medical School. Faculdade de Ciências Médicas. Universidade Nova de Lisboa. Lisboa. Portugal
| |
Collapse
|
18
|
Orwick A, Sears SM, Sharp CN, Doll MA, Shah PP, Beverly LJ, Siskind LJ. Lung cancer-kidney cross talk induces kidney injury, interstitial fibrosis, and enhances cisplatin-induced nephrotoxicity. Am J Physiol Renal Physiol 2023; 324:F287-F300. [PMID: 36727944 PMCID: PMC9988526 DOI: 10.1152/ajprenal.00317.2022] [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: 01/04/2023] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Patients with cancer represent a unique patient population with increased susceptibility to kidney disease. Drug-induced acute kidney injury (AKI) in patients with cancer is a common problem. Cisplatin is a highly effective treatment used in many solid-organ cancers and causes AKI in 30% of patients, increasing the risk of chronic kidney disease development. Most preclinical cisplatin toxicity studies have been completed in mice without cancer. We believe that the physiology of patients with cancer is not adequately represented in preclinical models, and the objective of this study was to determine how lung cancer will alter the nephrotoxicity of cisplatin. A genetically engineered mouse model and a syngeneic xenograft model of lung cancer were used. Mice were divided into the following four groups: 1) noncancer/vehicle, 2) noncancer/cisplatin, 3) cancer/vehicle, and 4) cancer/cisplatin. Mice were administered cisplatin via intraperitoneal injection once a week for 4 wk. Animals were euthanized 72 h following their final cisplatin injection. Mice with lung cancer had increased renal toxicity, injury, and fibrosis following repeated low doses of cisplatin. In addition, lung cancer alone induced kidney injury and fibrosis in the kidney before cisplatin treatment. In conclusion, this is the first study that we are aware of that assesses the impact of cancer on the kidney in conjunction with the nephrotoxicity of cisplatin. We believe that cancer is providing the first hit to the kidney and the subsequent damage from repeated doses of cisplatin becomes unsurmountable, leading to AKI and progression to chronic kidney disease.NEW & NOTEWORTHY Patients with cancer have impaired kidney function and increased susceptibility to nephrotoxic agents. Cisplatin is a commonly used chemotherapeutic with nephrotoxicity as the dose-limiting side effect. Cisplatin nephrotoxicity is almost exclusively studied in mice without cancer. Our current preclinical models do not adequately represent the complexity of patients with cancer. This study demonstrates increased renal toxicity, injury, and fibrosis in mice with lung cancer, which is exacerbated with cisplatin treatment. These results highlight the necessity of using preclinical models that more accurately capture the altered physiology of patients with cancer treated with cisplatin.
Collapse
Affiliation(s)
- Andrew Orwick
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Sophia M Sears
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Cierra N Sharp
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Mark A Doll
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Parag P Shah
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States
- Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States
| | - Levi J Beverly
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States
- Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
- Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States
| |
Collapse
|
19
|
Feng J, Li H, Wang S. Hydrogen sulfide alleviates uremic cardiomyopathy by regulating PI3K/PKB/mTOR-mediated overactive autophagy in 5/6 nephrectomy mice. Front Pharmacol 2022; 13:1027597. [PMID: 36588697 PMCID: PMC9797717 DOI: 10.3389/fphar.2022.1027597] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
The gasotransmitter hydrogen sulfide (H2S) plays important physiological and pathological roles in the cardiovascular system. However, the involvement of H2S in recovery from uremic cardiomyopathy (UCM) remains unclear. This study aimed to determine the therapeutic efficacy and elucidate the underlying mechanisms of H2S in UCM. A UCM model was established by 5/6 nephrectomy in 10-week-old C57BL/6 mice. Mice were treated with sodium hydrosulfide (NaHS, H2S donor), L-cysteine [L-Cys, cystathionine gamma-lyase (CSE) substrate], and propargylglycine (PPG, CSE inhibitor). Treatment of H9C2 cardiomyocytes utilized different concentrations of uremic serum, NaHS, PPG, and PI3K inhibitors (LY294002). Mouse heart function was assessed by echocardiography. Pathological changes in mouse myocardial tissue were identified using hematoxylin and eosin and Masson's trichrome staining. Cell viability was assessed using the Cell Counting Kit-8. The protein expressions of CSE, p-PI3K, PI3K, p-PKB, PKB, p-mTOR, mTOR, and autophagy-related markers (Beclin-1, P62, and LC3) were detected using Western blotting. We found that NaHS and L-Cys treatment attenuated myocardial disarray, fibrosis, and left ventricular dysfunction in UCM mice. These abnormalities were further aggravated by PPG supplementation. Enhanced autophagy and decreased phosphorylation of PI3K, PKB, and mTOR protein expression by UCM were altered by NaHS and L-Cys treatment. In vitro, uremic serum increased overactive autophagy and decreased the phosphorylation levels of PI3K, PKB, and mTOR in cardiomyocytes, which was substantially exacerbated by endogenous H2S deficiency and attenuated by pre-treatment with 100 µm NaHS. However, the protective effects of NaHS were completely inhibited by LY294002. These findings support a protective effect of H2S exerted against UCM by reducing overactive autophagy through activation of the PI3K/PKB/mTOR pathway.
Collapse
|
20
|
Kramer T, Brinkkoetter P, Rosenkranz S. Right Heart Function in Cardiorenal Syndrome. Curr Heart Fail Rep 2022; 19:386-399. [PMID: 36166185 PMCID: PMC9653308 DOI: 10.1007/s11897-022-00574-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW Since CRS is critically dependent on right heart function and involved in interorgan crosstalk, assessment and monitoring of both right heart and kidney function are of utmost importance for clinical outcomes. This systematic review aims to comprehensively report on novel diagnostic and therapeutic paradigms that are gaining importance for the clinical management of the growing heart failure population suffering from CRS. RECENT FINDINGS Cardiorenal syndrome (CRS) in patients with heart failure is associated with poor outcome. Although systemic venous congestion and elevated central venous pressure have been recognized as main contributors to CRS, they are often neglected in clinical practice. The delicate hemodynamic balance in CRS is particularly determined by the respective status of the right heart. The consideration of hemodynamic and CRS profiles is advantageous in tailoring treatment for better preservation of renal function. Assessment and monitoring of right heart and renal function by known and emerging tools like renal Doppler ultrasonography or new biomarkers may have direct clinical implications.
Collapse
Affiliation(s)
- Tilmann Kramer
- Klinik III Für Innere Medizin, Herzzentrum Der Universität Zu Köln, Köln, Germany.
- Cologne Cardiovascular Research Center (CCRC), Klinikum Der Universität Zu Köln, Köln, Germany.
| | - Paul Brinkkoetter
- Cologne Cardiovascular Research Center (CCRC), Klinikum Der Universität Zu Köln, Köln, Germany
- Klinik II Für Innere Medizin, Nephrologie, Universität Zu Köln, Köln, Germany
- Center for Molecular Medicine Cologne (CMMC), Universität Zu Köln, Köln, Germany
| | - Stephan Rosenkranz
- Klinik III Für Innere Medizin, Herzzentrum Der Universität Zu Köln, Köln, Germany
- Cologne Cardiovascular Research Center (CCRC), Klinikum Der Universität Zu Köln, Köln, Germany
- Center for Molecular Medicine Cologne (CMMC), Universität Zu Köln, Köln, Germany
| |
Collapse
|
21
|
Prokopidis K, Isanejad M, Akpan A, Stefil M, Tajik B, Giannos P, Venturelli M, Sankaranarayanan R. Exercise and nutritional interventions on sarcopenia and frailty in heart failure: a narrative review of systematic reviews and meta-analyses. ESC Heart Fail 2022; 9:2787-2799. [PMID: 35840310 PMCID: PMC9715780 DOI: 10.1002/ehf2.14052] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/16/2022] [Accepted: 06/27/2022] [Indexed: 11/10/2022] Open
Abstract
The purpose of this review is to describe the present evidence for exercise and nutritional interventions as potential contributors in the treatment of sarcopenia and frailty (i.e. muscle mass and physical function decline) and the risk of cardiorenal metabolic comorbidity in people with heart failure (HF). Evidence primarily from cross-sectional studies suggests that the prevalence of sarcopenia in people with HF is 37% for men and 33% for women, which contributes to cardiac cachexia, frailty, lower quality of life, and increased mortality rate. We explored the impact of resistance and aerobic exercise, and nutrition on measures of sarcopenia and frailty, and quality of life following the assessment of 35 systematic reviews and meta-analyses. The majority of clinical trials have focused on resistance, aerobic, and concurrent exercise to counteract the progressive loss of muscle mass and strength in people with HF, while promising effects have also been shown via utilization of vitamin D and iron supplementation by reducing tumour necrosis factor-alpha (TNF-a), c-reactive protein (CRP), and interleukin-6 (IL-6) levels. Experimental studies combining the concomitant effect of exercise and nutrition on measures of sarcopenia and frailty in people with HF are scarce. There is a pressing need for further research and well-designed clinical trials incorporating the anabolic and anti-catabolic effects of concurrent exercise and nutrition strategies in people with HF.
Collapse
Affiliation(s)
- Konstantinos Prokopidis
- Department of Musculoskeletal Biology, Institute of Life Course and Medical SciencesUniversity of LiverpoolLiverpoolUK
| | - Masoud Isanejad
- Department of Musculoskeletal Biology, Institute of Life Course and Medical SciencesUniversity of LiverpoolLiverpoolUK
| | - Asangaedem Akpan
- Department of Musculoskeletal Biology, Institute of Life Course and Medical SciencesUniversity of LiverpoolLiverpoolUK
- Aintree University HospitalLiverpool University Hospitals NHS Foundation TrustLiverpoolUK
| | - Maria Stefil
- Liverpool Centre for Cardiovascular ScienceUniversity of LiverpoolLiverpoolUK
- Department of Neurosciences, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Behnam Tajik
- Kuopio Musculoskeletal Research UnitUniversity of Eastern FinlandKuopioFinland
- National Institute for Health Research Northwest Coast CRNLiverpoolUK
| | - Panagiotis Giannos
- Department of Life Sciences, Faculty of Natural SciencesImperial College LondonLondonUK
| | - Massimo Venturelli
- Department of Neurosciences, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Rajiv Sankaranarayanan
- Liverpool Centre for Cardiovascular ScienceUniversity of LiverpoolLiverpoolUK
- Liverpool Heart and Chest Hospital NHS Foundation TrustLiverpoolUK
| |
Collapse
|
22
|
Pradhan SK, Adnani H, Safadi R, Yerigeri K, Nayak S, Raina R, Sinha R. Cardiorenal syndrome in the pediatric population: A systematic review. Ann Pediatr Cardiol 2022; 15:493-510. [PMID: 37152514 PMCID: PMC10158476 DOI: 10.4103/apc.apc_50_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/26/2022] [Accepted: 08/17/2022] [Indexed: 03/03/2023] Open
Abstract
The concept of cardiorenal syndrome (CRS) is derived from the crosstalk between the heart and kidneys in pathological conditions. Despite the rising importance of CRS, there is a paucity of information on the understanding of its pathophysiology and management, increasing both morbidity and mortality for patients. This review summarizes the existing conceptual pathophysiology of different types of CRS and delves into the associated therapeutic modalities with a focus on pediatric cases. Prospective or retrospective observational studies, comparative studies, case reports, case-control, and cross-sectional studies that include pediatric patients with CRS were included in this review. Literature was searched using PubMed, EMBASE, and Google Scholar with keywords including "cardio-renal syndrome, type," "reno-cardio syndrome," "children," "acute kidney injury," and "acute decompensated heart failure" from January 2000 to January 2021. A total of 14 pediatric studies were ultimately included and analyzed, comprising a combined population of 3608 children of which 32% had CRS. Of the 14 studies, 57% were based on type 1 CRS, 14% on types 2 and 3 CRS, and 7% were on types 4 and 5 CRS. The majority of included studies were prospective cohort, although a wide spectrum was observed in terms of patient age, comorbidities, etiologies, and treatment strategies. Commonly observed comorbidities in CRS type 1 were hematologic, oncologic, cardiology-related side effects, muscular dystrophy, and pneumonia/bronchiolitis. CRS, particularly type 1, is prevalent in children and has a significant risk of mortality. The current treatment regimen primarily involves diuretics, extracorporeal fluid removal, and treatment of underlying etiologies and comorbidities.
Collapse
Affiliation(s)
- Subal Kumar Pradhan
- Division of Pediatric Nephrology, Sardar Vallabhbhai Patel Post Graduate Institute of Pediatrics and SCB Medical College, Cuttack, Odisha, India
| | - Harsha Adnani
- Anne Arundel Medical Center, Luminis Health System, Annapolis, Maryland, USA
| | - Rama Safadi
- Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, Ohio, USA
| | - Keval Yerigeri
- Department of Nephrology, Akron, Ohio, USA, Children’s Hospital, Akron, Ohio, USA
| | - Snehamayee Nayak
- Department of Pediatrics, Sardar Vallabhbhai Patel Post Graduate Institute of Pediatrics and SCB Medical College, Cuttack, Odisha, India
| | - Rupesh Raina
- Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, Ohio, USA
- Department of Nephrology, Akron, Ohio, USA, Children’s Hospital, Akron, Ohio, USA
| | - Rajiv Sinha
- Division of Pediatric Nephrology, Institute of Child Health, Kolkata, West Bengal, India
- Department of Pediatrics, Apollo Gleneagles Hospital, Kolkata, West Bengal, India
| |
Collapse
|
23
|
Hernandez L, Laucyte-Cibulskiene A, Ward LJ, Kautzky-Willer A, Herrero MT, Norris CM, Raparelli V, Pilote L, Stenvinkel P, Kublickiene K. Gender dimension in cardio-pulmonary continuum. Front Cardiovasc Med 2022; 9:916194. [PMID: 36003909 PMCID: PMC9393639 DOI: 10.3389/fcvm.2022.916194] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
Cardio-pulmonary diseases, which were once regarded as a man's illness, have been one of the leading causes of morbidity and mortality for both men and women in many countries in recent years. Both gender and sex influence the functional and structural changes in the human body and therefore play an important role in disease clinical manifestation, treatment choice, and/or response to treatment and prognosis of health outcomes. The gender dimension integrates sex and gender analysis in health sciences and medical research, however, it is still relatively overlooked suggesting the need for empowerment in the medical research community. Latest advances in the field of cardiovascular research have provided supportive evidence that the application of biological variables of sex has led to the understanding that heart disease in females may have different pathophysiology compared to males, particularly in younger adults. It has also resulted in new diagnostic techniques and a better understanding of symptomatology, while gender analysis has informed more appropriate risk stratification and prevention strategies. The existing knowledge in the pulmonary field shows the higher prevalence of pulmonary disorders among females, however, the role of gender as a socio-cultural construct has yet to be explored for the implementation of targeted interventions. The purpose of this review is to introduce the concept of gender dimension and its importance for the cardiopulmonary continuum with a focus on shared pathophysiology and disease presentation in addition to interrelation with chronic kidney disease. The review presents basic knowledge of what gender dimension means, and the application of sex and gender aspects in cardiovascular medicine with a specific focus on early pulmonary development, pulmonary hypertension, and chronic obstructive pulmonary disease (COPD). Early vascular aging and inflammation have been presented as a potential pathophysiological link, with further interactions between the cardiopulmonary continuum and chronic kidney disease. Finally, implications for potential future research have been provided to increase the impact of gender dimension on research excellence that would add value to everybody, foster toward precision medicine and ultimately improve human health.
Collapse
Affiliation(s)
- Leah Hernandez
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Agne Laucyte-Cibulskiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Nephrology, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Liam J. Ward
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | - Alexandra Kautzky-Willer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Maria-Trinidad Herrero
- Clinical and Experimental Neuroscience, Institutes for Aging Research and Bio-Health Research of Murcia, School of Medicine, University of Murcia, Murcia, Spain
| | - Colleen M. Norris
- Faculty of Nursing, University of Alberta, Edmonton, AB, Canada
- Cardiovascular and Stroke Strategic Clinical Network, Alberta Health Services, Edmonton, AB, Canada
| | - Valeria Raparelli
- Faculty of Nursing, University of Alberta, Edmonton, AB, Canada
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- University Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy
| | - Louise Pilote
- Division of Clinical Epidemiology, Research Institute of McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
24
|
Muciño-Bermejo MJ. Extracorporeal organ support and the kidney. FRONTIERS IN NEPHROLOGY 2022; 2:924363. [PMID: 37674997 PMCID: PMC10479766 DOI: 10.3389/fneph.2022.924363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/01/2022] [Indexed: 09/08/2023]
Abstract
The concept of extracorporeal organ support (ECOS) encompasses kidney, respiratory, cardiac and hepatic support. In an era of increasing incidence and survival of patients with single or multiple organ failure, knowledge on both multiorgan crosstalk and the physiopathological consequences of extracorporeal organ support have become increasingly important. Immerse within the cross-talk of multiple organ failure (MOF), Acute kidney injury (AKI) may be a part of the clinical presentation in patients undergoing ECOS, either as a concurrent clinical issue since the very start of ECOS or as a de novo event at any point in the clinical course. At any point during the clinical course of a patient with single or multiple organ failure undergoing ECOS, renal function may improve or deteriorate, as a result of the interaction of multiple factors, including multiorgan crosstalk and physiological consequences of ECOS. Common physiopathological ways in which ECOS may influence renal function includes: 1) multiorgan crosstalk (preexisting or de-novo 2)Hemodynamic changes and 3) ECOS-associated coagulation abnormalities and 3) Also, cytokine profile switch, neurohumoral changes and toxins clearance may contribute to the expected physiological changes related to ECOS. The main objective of this review is to summarize the described mechanisms influencing the renal function during the course of ECOS, including renal replacement therapy, extracorporeal membrane oxygenation/carbon dioxide removal and albumin dialysis.
Collapse
Affiliation(s)
- Maria-Jimena Muciño-Bermejo
- Intensive Care Unit, The American British Cowdray Medical Center, Mexico City, Mexico
- International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy
- Health Sciences Department, Anahuac University, Mexico City, Mexico
- Medical Division, Medecins SansFontières – OCBA (Operational Centre Barcelona-Athens), Barcelona, Spain
| |
Collapse
|
25
|
Husain-Syed F, Reis T, Kashani K, Ronco C. Advances in laboratory detection of acute kidney injury. Pract Lab Med 2022; 31:e00283. [PMID: 35677313 PMCID: PMC9168173 DOI: 10.1016/j.plabm.2022.e00283] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/27/2022] [Accepted: 05/24/2022] [Indexed: 12/14/2022] Open
Abstract
Recent advances have improved our understanding of the epidemiology and pathophysiology of acute kidney injury (AKI). So far, the Kidney Disease: Improving Global Outcome guidelines define and stratify kidney injury based on increases in serum creatinine level and/or decreases in urine output. Although the term AKI acknowledges the existence of cellular injury, its diagnosis is still only defined by the reduced excretory function of the kidney. New biomarkers that aid a better understanding of the relationship between acute tubular injury and kidney dysfunction have been identified, reflecting the advances in molecular biology. The expression of some of these novel biomarkers precedes changes in conventional biomarkers or can increase their predictive power. Therefore, they might enhance the clinical accuracy of the definition of AKI. This review summarizes the limitations of the current AKI classification and a panel of candidate biomarkers for augmenting AKI classification and recognition of AKI subphenotypes. We expect that the integration of appropriately selected biomarkers in routine clinical practice can improve AKI care.
Collapse
Affiliation(s)
- Faeq Husain-Syed
- Department of Internal Medicine II, University Hospital Giessen and Marburg, Justus-Liebig-University Giessen, Klinikstraße 33, 35392, Giessen, Germany
| | - Thiago Reis
- Laboratory of Molecular Pharmacology, Faculty of Health Sciences, University of Brasília, Brasília, Distrito Federal, Brazil
- Department of Nephrology and Kidney Transplantation, Clínica de Doenças Renais de Brasília, DF Star Hospital, Rede D'Or São Luiz, Brasília, Distrito Federal, Brazil
| | - Kianoush Kashani
- Division of Nephrology and Hypertension, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Claudio Ronco
- Department of Medicine (DIMED), Università di Padova, Via Giustiniani, 2–35128, Padua, Italy
- International Renal Research Institute of Vicenza, Via Rodolfi, 37–36100, Vicenza, Italy
- Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, Via Rodolfi, 37–36100, Vicenza, Italy
| |
Collapse
|
26
|
Qiao J, Cui L. Multi-Omics Techniques Make it Possible to Analyze Sepsis-Associated Acute Kidney Injury Comprehensively. Front Immunol 2022; 13:905601. [PMID: 35874763 PMCID: PMC9300837 DOI: 10.3389/fimmu.2022.905601] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/10/2022] [Indexed: 12/29/2022] Open
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is a common complication in critically ill patients with high morbidity and mortality. SA-AKI varies considerably in disease presentation, progression, and response to treatment, highlighting the heterogeneity of the underlying biological mechanisms. In this review, we briefly describe the pathophysiology of SA-AKI, biomarkers, reference databases, and available omics techniques. Advances in omics technology allow for comprehensive analysis of SA-AKI, and the integration of multiple omics provides an opportunity to understand the information flow behind the disease. These approaches will drive a shift in current paradigms for the prevention, diagnosis, and staging and provide the renal community with significant advances in precision medicine in SA-AKI analysis.
Collapse
Affiliation(s)
- Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, China
- *Correspondence: Liyan Cui,
| |
Collapse
|
27
|
In-hospital mortality of critically Ill patients with interactions of acute kidney injury and acute respiratory failure in the resource-limited settings: Results from SEA-AKI study. J Crit Care 2022; 71:154103. [PMID: 35779395 DOI: 10.1016/j.jcrc.2022.154103] [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: 02/06/2022] [Revised: 05/31/2022] [Accepted: 06/11/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE Our goal was to describe clinical outcomes and explore the physiological interactions between acute kidney injury (AKI) and acute respiratory failure (ARF) in critically ill patients. MATERIALS AND METHODS Data were retrieved from the SEA-AKI study, a multinational multicenter database of adult ICUs from Thailand, Laos, and Indonesia. AKI was defined using KDIGO criteria stage 2-3. ARF was defined by being mechanically ventilated. Patients were assigned into 6 patterns based on AKI and ARF sequence: "no AKI/ARF", "ARF alone", "AKI alone", "ARF first", "AKI first", and "Concurrent AKI-ARF". The primary outcome was in-hospital mortality of each pattern. RESULTS A final cohort of 5468 patients were eligible for the analysis. The "Concurrent AKI-ARF" had the highest in-hospital mortality of 69.6%. The "AKI first" and the "ARF first" had in-hospital mortality of 54.4% and 53%, respectively. Among patients with single organ failure, in-hospital mortality was 14.6% and 31.5% in the "AKI alone" and the "ARF alone", accordingly. In-hospital mortality was 12.4% in patients without AKI and ARF. CONCLUSION Critically ill patients with ARF and AKI are at higher risk of in-hospital death. Different patterns of AKI and ARF interaction result in unique clinical outcomes as well as risk factors.
Collapse
|
28
|
Baig SH, Vaid U, Yoo EJ. The Impact of Chronic Medical Conditions on Mortality in Acute Respiratory Distress Syndrome. J Intensive Care Med 2022; 38:78-85. [PMID: 35722731 DOI: 10.1177/08850666221108079] [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: 11/17/2022]
Abstract
PURPOSE To examine the impact of chronic comorbidities on mortality in Acute Respiratory Distress Syndrome (ARDS). MATERIALS AND METHODS Retrospective cohort study of adults with ARDS (ICD-10-CM code J80) from the National Inpatient Sample between January, 2016 and December, 2018. For the primary outcome of mortality, we conducted weighted logistic regression adjusting for factors identified on univariate analysis as potentially significant or differing between the two groups at baseline. We used negative binomial regression adjusting for the same comorbidities to identify risk factors for longer length of stay (LOS) among ARDS survivors. RESULTS After exclusions, 1046 records were analyzed (3355 ARDS survivors and 1875 non-survivors.) The comorbidities examined included hypertension, diabetes mellitus, obesity, hypothyroidism, alcohol and drug use, chronic kidney disease (CKD), cardiovascular disease, chronic liver disease, chronic pulmonary disease and malignancy. In multivariate analysis, we found that malignancy (OR 2.26, 95% CI 1.84-2.78, p < 0.001), cardiovascular disease (OR 1.54, 95% CI 1.23-1.92, p < 0.001), and CKD (OR 1.75, 95% CI 1.22-2.50, p = 0.002) increased the risk of death. In interaction analyses, cardiovascular disease combined with either malignancy or CKD conferred higher odds of death compared to either risk factor alone. CONCLUSIONS The comorbidity of malignancy confers the most reliable risk of poor outcomes in ARDS with higher odds of hospital death and a simultaneous association with longer hospital LOS among survivors.
Collapse
Affiliation(s)
- Saqib H Baig
- Division of Pulmonary, Allergy and Critical Care Medicine, Jane and Leonard Korman Respiratory Institute, 12313Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Urvashi Vaid
- Division of Pulmonary, Allergy and Critical Care Medicine, Jane and Leonard Korman Respiratory Institute, 12313Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Erika J Yoo
- Division of Pulmonary, Allergy and Critical Care Medicine, Jane and Leonard Korman Respiratory Institute, 12313Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
29
|
Gabbin B, Meraviglia V, Mummery CL, Rabelink TJ, van Meer BJ, van den Berg CW, Bellin M. Toward Human Models of Cardiorenal Syndrome in vitro. Front Cardiovasc Med 2022; 9:889553. [PMID: 35694669 PMCID: PMC9177996 DOI: 10.3389/fcvm.2022.889553] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Heart and kidney diseases cause high morbidity and mortality. Heart and kidneys have vital functions in the human body and, interestingly, reciprocally influence each other’s behavior: pathological changes in one organ can damage the other. Cardiorenal syndrome (CRS) is a group of disorders in which there is combined dysfunction of both heart and kidney, but its underlying biological mechanisms are not fully understood. This is because complex, multifactorial, and dynamic mechanisms are likely involved. Effective treatments are currently unavailable, but this may be resolved if more was known about how the disease develops and progresses. To date, CRS has actually only been modeled in mice and rats in vivo. Even though these models can capture cardiorenal interaction, they are difficult to manipulate and control. Moreover, interspecies differences may limit extrapolation to patients. The questions we address here are what would it take to model CRS in vitro and how far are we? There are already multiple independent in vitro (human) models of heart and kidney, but none have so far captured their dynamic organ-organ crosstalk. Advanced in vitro human models can provide an insight in disease mechanisms and offer a platform for therapy development. CRS represents an exemplary disease illustrating the need to develop more complex models to study organ-organ interaction in-a-dish. Human induced pluripotent stem cells in combination with microfluidic chips are one powerful tool with potential to recapitulate the characteristics of CRS in vitro. In this review, we provide an overview of the existing in vivo and in vitro models to study CRS, their limitations and new perspectives on how heart-kidney physiological and pathological interaction could be investigated in vitro for future applications.
Collapse
Affiliation(s)
- Beatrice Gabbin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Enschede, Netherlands
| | - Ton J. Rabelink
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Berend J. van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Cathelijne W. van den Berg
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department of Biology, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
- *Correspondence: Milena Bellin, ,
| |
Collapse
|
30
|
Rhodes CJ, Wharton J, Swietlik EM, Harbaum L, Girerd B, Coghlan JG, Lordan J, Church C, Pepke-Zaba J, Toshner M, Wort SJ, Kiely DG, Condliffe R, Lawrie A, Gräf S, Montani D, Boucly A, Sitbon O, Humbert M, Howard LS, Morrell NW, Wilkins MR. Using the Plasma Proteome for Risk Stratifying Patients with Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2022; 205:1102-1111. [PMID: 35081018 PMCID: PMC9851485 DOI: 10.1164/rccm.202105-1118oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Rationale: NT-proBNP (N-terminal pro-brain natriuretic peptide), a biomarker of cardiac origin, is used to risk stratify patients with pulmonary arterial hypertension (PAH). Its limitations include poor sensitivity to early vascular pathology. Other biomarkers of vascular or systemic origin may also be useful in the management of PAH. Objectives: Identify prognostic proteins in PAH that complement NT-proBNP and clinical risk scores. Methods: An aptamer-based assay (SomaScan version 4) targeting 4,152 proteins was used to measure plasma proteins in patients with idiopathic, heritable, or drug-induced PAH from the UK National Cohort of PAH (n = 357) and the French EFORT (Evaluation of Prognostic Factors and Therapeutic Targets in PAH) study (n = 79). Prognostic proteins were identified in discovery-replication analyses of UK samples. Proteins independent of 6-minute-walk distance and NT-proBNP entered least absolute shrinkage and selection operator modeling, and the best combination in a single score was evaluated against clinical targets in EFORT. Measurements and Main Results: Thirty-one proteins robustly informed prognosis independent of NT-proBNP and 6-minute-walk distance in the UK cohort. A weighted combination score of six proteins was validated at baseline (5-yr mortality; area under the curve [AUC], 0.73; 95% confidence interval [CI], 0.63-0.85) and follow-up in EFORT (AUC, 0.84; 95% CI, 0.75-0.94; P = 9.96 × 10-6). The protein score risk stratified patients independent of established clinical targets and risk equations. The addition of the six-protein model score to NT-proBNP improved prediction of 5-year outcomes from AUC 0.762 (0.702-0.821) to 0.818 (0.767-0.869) by receiver operating characteristic analysis (P = 0.00426 for difference in AUC) in the UK replication and French samples combined. Conclusions: The plasma proteome informs prognosis beyond established factors in PAH and may provide a more sensitive measure of therapeutic response.
Collapse
Affiliation(s)
- Christopher J Rhodes
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Emilia M Swietlik
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Lars Harbaum
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Barbara Girerd
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - J Gerry Coghlan
- Department of Cardiology, Royal Free Campus, University College London, London, United Kingdom
| | - James Lordan
- University of Newcastle upon Tyne, Newcastle upon Tyne, United Kingdom
| | - Colin Church
- University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Joanna Pepke-Zaba
- Pulmonary Vascular Disease Unit, Royal Papworth Hospital, Cambridge, United Kingdom
| | - Mark Toshner
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Stephen J Wort
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - David G Kiely
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom.,Sheffield Pulmonary Vascular Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom; and
| | - Robin Condliffe
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom.,Sheffield Pulmonary Vascular Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom; and
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,BioResource for Translational Research, National Institute for Health Research Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - David Montani
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Athénaïs Boucly
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Olivier Sitbon
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Marc Humbert
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Luke S Howard
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | |
Collapse
|
31
|
Rachel M, Galiniak S, Biesiadecki M, Gala-Błądzińska A. Renal Function in Patients with Cystic Fibrosis: A Single-Center Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:5454. [PMID: 35564849 PMCID: PMC9105043 DOI: 10.3390/ijerph19095454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023]
Abstract
Cystic fibrosis (CF) is the most common incurable autosomal recessive disease affecting the Caucasian population. As the prognosis for life extension of CF patients improves, co-morbidities, including kidney disease, become more common. Identifying those at the highest risk of kidney injury is therefore extremely important. The aim of this study was to evaluate the biomarkers of renal function in 50 CF patients using the estimated glomerular filtration rate (eGFR) based on creatinine and cystatin C equation as well as serum creatinine (sCr), serum cystatin C (CysC), serum urea and urinary neutrophil gelatinase-associated lipocalin (uNGAL) concentrations. sCr, CysC, urea and uNGAL were estimated. eGFR was calculated according to the CKD-EPI formula. CysC was significantly increased, while eGFR was significantly lower in the CF group than in the controls (p < 0.001 and p < 0.01, respectively). There was no significant difference in the sCr, urea and uNGAL concentrations between patients with CF and healthy subjects. For the purpose of our analysis, in order to assess renal function in patients with CF in clinical practice, the concentration of serum CysC and eGFRCKD-EPI should be determined. Patients with CF presented with renal function impairment pictured by increased serum CysC and decreased eGFR values compared to controls. Unchanged uNGAL concentrations suggested preserved tubular function despite aminoglycoside treatment. Further prospective studies are needed to clarify whether kidney impairment observed in the course of CF progresses.
Collapse
Affiliation(s)
- Marta Rachel
- Department of Allergology, Provincial Hospital No. 2, Lwowska 60, 35-301 Rzeszów, Poland
- Institute of Medical Sciences, Medical College of Rzeszów University, Rzeszów University, Warzywna 1a, 35-310 Rzeszów, Poland; (M.B.); (A.G.-B.)
| | - Sabina Galiniak
- Institute of Medical Sciences, Medical College of Rzeszów University, Rzeszów University, Warzywna 1a, 35-310 Rzeszów, Poland; (M.B.); (A.G.-B.)
| | - Marek Biesiadecki
- Institute of Medical Sciences, Medical College of Rzeszów University, Rzeszów University, Warzywna 1a, 35-310 Rzeszów, Poland; (M.B.); (A.G.-B.)
| | - Agnieszka Gala-Błądzińska
- Institute of Medical Sciences, Medical College of Rzeszów University, Rzeszów University, Warzywna 1a, 35-310 Rzeszów, Poland; (M.B.); (A.G.-B.)
- Department of Internal Medicine, Nephrology and Endocrinology, Provincial Hospital No. 2, Lwowska 60, 35-301 Rzeszów, Poland
| |
Collapse
|
32
|
Liang HY, Hsiao YL, Yeh HC, Ting IW, Lin CC, Chiang HY, Kuo CC. Associations Between Myocardial Diastolic Dysfunction and Cardiovascular Mortality in Chronic Kidney Disease: A Large Single-Center Cohort Study. J Am Soc Echocardiogr 2021; 35:395-407. [PMID: 34915133 DOI: 10.1016/j.echo.2021.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND The clinical burden and prognostic role of diastolic dysfunction (DD), based on the latest American Society of Echocardiography (ASE) 2016 guidelines, remains unclear in patients with chronic kidney disease (CKD). Moreover, risk mapping of concomitant systolic dysfunction and DD to evaluate the hazard of cardiovascular (CV) mortality in CKD remains unexplored. METHODS This retrospective cohort study identified 20,257 adult patients who underwent comprehensive echocardiography between 2008 and 2016 at a tertiary medical center in central Taiwan. The patients were stratified by CKD stage, and the 3-year CV mortality risk in each CKD stratum was estimated through multivariable Cox proportional hazard modeling using left ventricular ejection fraction (LVEF) and DD grades based on ASE 2016 guideline as the main risk factors. RESULTS Compared with patients with CKD stages 1 and 2, those with CKD stages 4 and 5 had a significantly lower LVEF and more severe DD. Both LVEF (LVEF < 40% vs. ≥ 60%; adjusted hazard ratio [aHR] 3.17, 95% confidence interval [CI] 2.54-3.97) and DD grade (severe DD vs. normal diastolic function; aHR 3.33, 95% CI 2.33-4.76) were independently associated with 3-year CV mortality in the entire study population and had comparable effect sizes. The corresponding aHRs further increased to 4.20 (2.45-7.21) and 4.54 (2.20-9.38) in patients with CKD stages 4 and 5. Systolic and diastolic dysfunction demonstrated mutually augmentative effects on CV mortality. CONCLUSIONS These findings suggest that the current practice of cardioprotection for patients with CKD should be prioritized at an early stage along with conventional nephroprotection.
Collapse
Affiliation(s)
- Hsin-Yueh Liang
- Division of Cardiology, Department of Internal Medicine, China Medical University Hospital and College of Medicine, China Medical University, Taichung, Taiwan; Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Ya-Luan Hsiao
- Department of Health Policy and Management, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Hung-Chieh Yeh
- Division of Nephrology, Department of Internal Medicine, China Medical University Hospital and College of Medicine, China Medical University, Taichung, Taiwan; Big Data Center, China Medical University Hospital and College of Medicine, China Medical University, Taichung, Taiwan
| | - I-Wen Ting
- Division of Nephrology, Department of Internal Medicine, China Medical University Hospital and College of Medicine, China Medical University, Taichung, Taiwan; Big Data Center, China Medical University Hospital and College of Medicine, China Medical University, Taichung, Taiwan
| | - Che-Chen Lin
- Big Data Center, China Medical University Hospital and College of Medicine, China Medical University, Taichung, Taiwan
| | - Hsiu-Yin Chiang
- Big Data Center, China Medical University Hospital and College of Medicine, China Medical University, Taichung, Taiwan
| | - Chin-Chi Kuo
- Division of Nephrology, Department of Internal Medicine, China Medical University Hospital and College of Medicine, China Medical University, Taichung, Taiwan; Big Data Center, China Medical University Hospital and College of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
33
|
Ciccarelli M, Dawson D, Falcao-Pires I, Giacca M, Hamdani N, Heymans S, Hooghiemstra A, Leeuwis A, Hermkens D, Tocchetti CG, van der Velden J, Zacchigna S, Thum T. Reciprocal organ interactions during heart failure: a position paper from the ESC Working Group on Myocardial Function. Cardiovasc Res 2021; 117:2416-2433. [PMID: 33483724 PMCID: PMC8562335 DOI: 10.1093/cvr/cvab009] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/20/2021] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Heart failure-either with reduced or preserved ejection fraction (HFrEF/HFpEF)-is a clinical syndrome of multifactorial and gender-dependent aetiology, indicating the insufficiency of the heart to pump blood adequately to maintain blood flow to meet the body's needs. Typical symptoms commonly include shortness of breath, excessive fatigue with impaired exercise capacity, and peripheral oedema, thereby alluding to the fact that heart failure is a syndrome that affects multiple organ systems. Patients suffering from progressed heart failure have a very limited life expectancy, lower than that of numerous cancer types. In this position paper, we provide an overview regarding interactions between the heart and other organ systems, the clinical evidence, underlying mechanisms, potential available or yet-to-establish animal models to study such interactions and finally discuss potential new drug interventions to be developed in the future. Our working group suggests that more experimental research is required to understand the individual molecular mechanisms underlying heart failure and reinforces the urgency for tailored therapeutic interventions that target not only the heart but also other related affected organ systems to effectively treat heart failure as a clinical syndrome that affects and involves multiple organs.
Collapse
Affiliation(s)
- Michele Ciccarelli
- University of Salerno, Department of Medicine, Surgery and Dentistry, Via S. Allende 1, 84081, Baronissi(Salerno), Italy
| | - Dana Dawson
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2DZ, UK
| | - Inês Falcao-Pires
- Department of Surgery and Physiology, Cardiovascular Research and Development Center, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Mauro Giacca
- King’s College London, Molecular Medicine Laboratory, 125 Caldharbour Lane, London WC2R2LS, United Kingdom
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149 Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34129 Trieste, Italy
| | - Nazha Hamdani
- Department of Clinical Pharmacology and Molecular Cardiology, Institute of Physiology, Ruhr University Bochum, Universitätsstraße 150, D-44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Universitätsstraße 150, D-44801 Bochum, Germany
| | - Stéphane Heymans
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, Bus 911, 3000 Leuven, Belgium
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
- ICIN-Netherlands Heart Institute, Holland Heart House, Moreelsepark 1, 3511 EP Utrecht, the Netherlands
| | - Astrid Hooghiemstra
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081HZ, Amsterdam, The Netherlands
- Department of Medical Humanities, Amsterdam Public Health Research Institute, Amsterdam UMC, Location VUmc, De Boelelaan 1089a, 1081HV, Amsterdam, The Netherlands
| | - Annebet Leeuwis
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081HZ, Amsterdam, The Netherlands
| | - Dorien Hermkens
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, the Netherlands
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
| | - Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Physiology, Amsterdam Cardiovascular Sciences, De Boelelaan 1118, 1081HZ Amsterdam, the Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34129 Trieste, Italy
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149 Trieste, Italy
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
- REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
- Fraunhofer Institute of Toxicology and Experimental Medicine, Nicolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| |
Collapse
|
34
|
Patel M, Bellanti F, Daryani NM, Noormohamed N, Hilbert DW, Young K, Kulkarni P, Copalu W, Gheyas F, Rizk ML. Population pharmacokinetic/pharmacodynamic assessment of imipenem/cilastatin/relebactam in patients with hospital-acquired/ventilator-associated bacterial pneumonia. Clin Transl Sci 2021; 15:396-408. [PMID: 34704389 PMCID: PMC8841461 DOI: 10.1111/cts.13158] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/30/2021] [Accepted: 08/29/2021] [Indexed: 01/01/2023] Open
Abstract
In the phase III RESTORE-IMI 2 study (ClinicalTrials.gov: NCT02493764), the combination antibacterial agent imipenem/cilastatin/relebactam (IMI/REL) demonstrated noninferiority to piperacillin/tazobactam for the end points of all-cause mortality at day 28 and favorable clinical response at the early follow-up visit in adult participants with gram-negative hospital-acquired bacterial pneumonia/ventilator-associated bacterial pneumonia (HABP/VABP). Existing population pharmacokinetic models for imipenem (IPM) and REL were updated using data from patients with HABP/VABP from RESTORE-IMI 2. Creatinine clearance (CrCl), body weight, infection type, and ventilation status were significant covariates in the updated model. The following simulations were performed to calculate the pharmacokinetic/pharmacodynamic joint probability of target attainment among patients with HABP/VABP and varying degrees of renal function: augmented renal clearance (CrCl ≥150 ml/min), normal renal function (CrCl ≥90 to <150 ml/min), renal impairment (mild, CrCl ≥60 to <90 ml/min; moderate, CrCl ≥30 to <60 ml/min; or severe, CrCl ≥15 to <30 ml/min), and end-stage renal disease (CrCl <15 ml/min). At the recommended IMI/REL dosing regimens across renal categories, greater than 90% of patients in all renal function groups were predicted to achieve joint pharmacokinetic/pharmacodynamic targets at a minimum inhibitory concentration breakpoint of ≤2 μg/ml, regardless of ventilation status. This modeling and simulation analysis supports use of the recommended IMI/REL dosing regimens, adjusted based on renal function, in patients with HABP/VABP.
Collapse
|
35
|
Rubio-Gracia J, Josa-Laorden C, Sánchez-Marteles M, Giménez-López I, Garcés Horna V, Morales Rull JL, Pérez-Calvo JI. Prognostic value of malnutrition in patients with acute heart failure and its influence on the interpretation of markers of systemic venous congestion. Med Clin (Barc) 2021; 157:371-379. [PMID: 33309049 DOI: 10.1016/j.medcli.2020.06.066] [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: 04/28/2020] [Revised: 06/21/2020] [Accepted: 06/24/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Malnutrition is frequent in patients with heart failure (HF) and contributes to increased systemic congestion, but also hinders its correct assessment, especially during decompensations. Estimating the degree of malnutrition and its relationship with systemic congestion is important to optimize treatment during decompensations. MATERIAL AND METHODS Retrospective cohort study in patients with acute HF. The population was stratified according to CONUT (Controlling Nutritional Status) and PNI (Prognostic Nutritional Index) nutrition indices in order to analyse their relationship with objective parameters of congestion and the prognostic value of malnutrition. RESULTS 309 patients were included. More than half presented some degree of malnutrition upon admission. The degree of congestion was significantly higher in malnourished patients, with a higher proportion of «comet tail artifacts» and a higher relative plasma volume. NT-proBNP concentrations, both on admission and at discharge, were also significantly higher in malnourished patients, regardless of the scale used. The univariate analysis identified the CONUT and PNI index as factors associated with one-year mortality from any cause (HR 1.62 [1.22-2.14]; p = 0.001) and PNI (HR 65 [0.53-0.80]; p = < 0.001), respectively. CONCLUSIONS A higher degree of malnutrition (determined by means of the CONUT and PNI indices) in patients with acute HF was associated with a higher presence of objective parameters of congestion and a higher one-year all-cause mortality.
Collapse
Affiliation(s)
- Jorge Rubio-Gracia
- Servicio de Medicina Interna, Hospital Clínico Universitario «Lozano Blesa», Zaragoza, España; Instituto de Investigación Sanitaria de Aragón (IIS), Zaragoza, España.
| | - Claudia Josa-Laorden
- Servicio de Medicina Interna, Hospital Clínico Universitario «Lozano Blesa», Zaragoza, España; Instituto de Investigación Sanitaria de Aragón (IIS), Zaragoza, España
| | - Marta Sánchez-Marteles
- Servicio de Medicina Interna, Hospital Clínico Universitario «Lozano Blesa», Zaragoza, España; Instituto de Investigación Sanitaria de Aragón (IIS), Zaragoza, España
| | - Ignacio Giménez-López
- Instituto de Investigación Sanitaria de Aragón (IIS), Zaragoza, España; Facultad de Medicina, Universidad de Zaragoza, Zaragoza, España; Instituto Aragonés de Ciencias de la Salud, Zaragoza, España
| | - Vanesa Garcés Horna
- Servicio de Medicina Interna, Hospital Clínico Universitario «Lozano Blesa», Zaragoza, España; Instituto de Investigación Sanitaria de Aragón (IIS), Zaragoza, España
| | | | - Juan Ignacio Pérez-Calvo
- Servicio de Medicina Interna, Hospital Clínico Universitario «Lozano Blesa», Zaragoza, España; Instituto de Investigación Sanitaria de Aragón (IIS), Zaragoza, España; Facultad de Medicina, Universidad de Zaragoza, Zaragoza, España
| |
Collapse
|
36
|
Demirel M, Külahçıoğlu Ş, Tokgöz HC, Akbal ÖY, Hakgör A, Karagöz A, Tanyeri S, Keskin B, Kültürsay B, Efe SÇ, Bayram Z, Tanboğa İH, Özdemir N, Kaymaz C. Impaired endothelium-dependent and endothelium-independent systemic vasodilatory reserve in pulmonary hypertension regardless the clinical group: A generalized dysfunction beyond the pulmonary arteries? Anatol J Cardiol 2021; 25:733-740. [PMID: 34622788 DOI: 10.5152/anatoljcardiol.2021.474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE Endothelium-dependent (ED) and endothelium-independent (EI) flow-mediated vasodilatation (FMD) have been used as measures of systemic arterial vasodilatory reserve. In this study, we aimed to assess both ED-FMD and EI-FMD in different groups with pulmonary hypertension (PH), and to investigate the relationship of these measures with clinical, echocardiographic, and invasive parameters of diseases severity and targeted treatment status. METHODS Our study population comprised 41 patients with PH [28 (68.2%) women, age 46.3±19.6 years] including idiopathic pulmonary arterial hypertension, Eisenmenger syndrome, and chronic thromboembolic PH in whom diagnosis were confirmed in accordance with current guidelines and 17 age and sex-matched healthy controls. The brachial artery (BA) was used for assessment of FMD with Duplex ultrasound, and serial changes in diameter were recorded at baseline, 1, and 3 minutes after termination of 2-minute external occlusive compression for ED-FMD, and after sublingual intake of glycerol trinitrate for EI-FMD, respectively. RESULTS Compared with controls, overall the PH group showed significantly lower ED-FMD (0.65±0.21 vs. 0.30±0.23 and 0.65±0.18 vs. 0.24±0.21) and EI-FMD (0.67±0.15 vs. 0.37±0.25 and 0.75±0.20 vs. 0.32±0.24) responses at 1st and 3rd min (p<0.001 for all). All these changes in the values of ED-FMD and EI-FMD were comparable among the PH subgroups. Neither ED-FMD nor EI-FMD were correlated with measures of PH severity and targeted therapy (TT) status (p>0.05). CONCLUSION Our results suggest an impaired BA vasodilatory reserve in patients with PH regardless of the clinical subgroup. Although these findings seem to be consistent with systemic dysfunction, acute FMD may not reflect the severity of PH and cannot be used as a potential surrogate for outcome in this setting.
Collapse
Affiliation(s)
- Muhittin Demirel
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Şeyhmus Külahçıoğlu
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Hacer Ceren Tokgöz
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Özgür Y Akbal
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Aykun Hakgör
- Department of Cardiology, Bingöl State Hospital; Bingöl-Turkey
| | - Ali Karagöz
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Seda Tanyeri
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Berhan Keskin
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Barkın Kültürsay
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Süleyman Çağan Efe
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Zübeyde Bayram
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | | | - Nihal Özdemir
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Cihangir Kaymaz
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| |
Collapse
|
37
|
Demirel M, Külahçıoğlu Ş, Tokgöz HC, Akbal ÖY, Hakgör A, Karagöz A, Tanyeri S, Keskin B, Kültürsay B, Efe SÇ, Bayram Z, Tanboğa İH, Özdemir N, Kaymaz C. Impaired endothelium-dependent and endothelium-independent systemic vasodilatory reserve in pulmonary hypertension regardless the clinical group: A generalized dysfunction beyond the pulmonary arteries? Anatol J Cardiol 2021. [PMID: 34622788 DOI: 10.5152/anatoljcardiol.2021.474)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE Endothelium-dependent (ED) and endothelium-independent (EI) flow-mediated vasodilatation (FMD) have been used as measures of systemic arterial vasodilatory reserve. In this study, we aimed to assess both ED-FMD and EI-FMD in different groups with pulmonary hypertension (PH), and to investigate the relationship of these measures with clinical, echocardiographic, and invasive parameters of diseases severity and targeted treatment status. METHODS Our study population comprised 41 patients with PH [28 (68.2%) women, age 46.3±19.6 years] including idiopathic pulmonary arterial hypertension, Eisenmenger syndrome, and chronic thromboembolic PH in whom diagnosis were confirmed in accordance with current guidelines and 17 age and sex-matched healthy controls. The brachial artery (BA) was used for assessment of FMD with Duplex ultrasound, and serial changes in diameter were recorded at baseline, 1, and 3 minutes after termination of 2-minute external occlusive compression for ED-FMD, and after sublingual intake of glycerol trinitrate for EI-FMD, respectively. RESULTS Compared with controls, overall the PH group showed significantly lower ED-FMD (0.65±0.21 vs. 0.30±0.23 and 0.65±0.18 vs. 0.24±0.21) and EI-FMD (0.67±0.15 vs. 0.37±0.25 and 0.75±0.20 vs. 0.32±0.24) responses at 1st and 3rd min (p<0.001 for all). All these changes in the values of ED-FMD and EI-FMD were comparable among the PH subgroups. Neither ED-FMD nor EI-FMD were correlated with measures of PH severity and targeted therapy (TT) status (p>0.05). CONCLUSION Our results suggest an impaired BA vasodilatory reserve in patients with PH regardless of the clinical subgroup. Although these findings seem to be consistent with systemic dysfunction, acute FMD may not reflect the severity of PH and cannot be used as a potential surrogate for outcome in this setting.
Collapse
Affiliation(s)
- Muhittin Demirel
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Şeyhmus Külahçıoğlu
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Hacer Ceren Tokgöz
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Özgür Y Akbal
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Aykun Hakgör
- Department of Cardiology, Bingöl State Hospital; Bingöl-Turkey
| | - Ali Karagöz
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Seda Tanyeri
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Berhan Keskin
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Barkın Kültürsay
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Süleyman Çağan Efe
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Zübeyde Bayram
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | | | - Nihal Özdemir
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| | - Cihangir Kaymaz
- Department of Cardiology, University of Health Sciences, Hamidiye Faculty of Medicine, Koşuyolu Yüksek İhtisas Training and Research Hospital; İstanbul-Turkey
| |
Collapse
|
38
|
Ge H, Liang Y, Fang Y, Jin Y, Su W, Zhang G, Wang J, Xiong H, Shang D, Chai Y, Liu Z, Wei H, Wang H, Zhang W, Ma F, Zhao W, Sun L, Huang H, Ma Q. Predictors of acute kidney injury in patients with acute decompensated heart failure in emergency departments in China. J Int Med Res 2021; 49:3000605211016208. [PMID: 34510958 PMCID: PMC8442502 DOI: 10.1177/03000605211016208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Objective This retrospective multicentre observational study was performed to assess
the predictors of acute kidney injury (AKI) in patients with acute
decompensated heart failure (ADHF) in emergency departments in China. Methods In total, 1743 consecutive patients with ADHF were recruited from August 2017
to January 2018. Clinical characteristics and outcomes were compared between
patients with and without AKI. Predictors of AKI occurrence and
underdiagnosis were assessed in multivariate regression analyses. Results Of the 1743 patients, 593 (34.0%) had AKI. AKI was partly associated with
short-term all-cause mortality and cost. Cardiovascular comorbidities such
as coronary heart disease, diabetes mellitus, and hypertension remained
significant predictors of AKI in the univariate analysis. AKI was
significantly more likely to occur in patients with a lower arterial pH,
lower albumin concentration, higher creatinine concentration, and higher
N-terminal pro-brain natriuretic peptide (NT-proBNP) concentration. Patients
treated with inotropic agents were significantly more likely to develop AKI
during their hospital stay. Conclusion This study suggests that cardiovascular comorbidities, arterial pH, the
albumin concentration, the creatinine concentration, the NT-proBNP
concentration, and use of inotropic agents are predictors of AKI in patients
with ADHF.
Collapse
Affiliation(s)
- Hongxia Ge
- Emergency Department, Peking University Third Hospital, No. 49 North Garden Road, Hai-dian District, Beijing, China
| | - Yang Liang
- Emergency Department, Peking University Third Hospital, No. 49 North Garden Road, Hai-dian District, Beijing, China
| | - Yingying Fang
- Emergency Department, Peking University Third Hospital, No. 49 North Garden Road, Hai-dian District, Beijing, China
| | - Yi Jin
- Emergency Department, Peking University Third Hospital, No. 49 North Garden Road, Hai-dian District, Beijing, China
| | - Wenting Su
- Emergency Department, Peking University Third Hospital, No. 49 North Garden Road, Hai-dian District, Beijing, China
| | - Guoqiang Zhang
- Emergency Department, China-Japan Friendship Hospital, Beijing, China
| | - Jing Wang
- Emergency Department, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Hui Xiong
- Emergency Department, Peking University First Hospital, Beijing, China
| | - Deya Shang
- Emergency Department, Shandong Provincial Hospital, Jinan, Shandong, China
| | - Yanfen Chai
- Emergency Department, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhi Liu
- Emergency Department, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hongyan Wei
- Emergency Department, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hairong Wang
- Emergency Department, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Zhang
- Emergency Department, Tianjin Third Central Hospital, Tianjin, China
| | - Fei Ma
- Emergency Department, Guangdong Provincial People's Hospital, Guangzhou, Guangdong, China
| | - Wei Zhao
- Emergency Department, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Li Sun
- Emergency Department, Shanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Huan Huang
- Emergency Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qingbian Ma
- Emergency Department, Peking University Third Hospital, No. 49 North Garden Road, Hai-dian District, Beijing, China
| |
Collapse
|
39
|
Ischemic preconditioning protects the heart against ischemia-reperfusion injury in chronic kidney disease in both males and females. Biol Sex Differ 2021; 12:49. [PMID: 34488888 PMCID: PMC8420010 DOI: 10.1186/s13293-021-00392-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Uremic cardiomyopathy is a common cardiovascular complication of chronic kidney disease (CKD) characterized by left ventricular hypertrophy (LVH) and fibrosis enhancing the susceptibility of the heart to acute myocardial infarction. In the early stages of CKD, approximately 60% of patients are women. We aimed to investigate the influence of sex on the severity of uremic cardiomyopathy and the infarct size-limiting effect of ischemic preconditioning (IPRE) in experimental CKD. METHODS CKD was induced by 5/6 nephrectomy in 9-week-old male and female Wistar rats. Two months later, serum and urine laboratory parameters were measured to verify the development of CKD. Transthoracic echocardiography was performed to assess cardiac function and morphology. Cardiomyocyte hypertrophy and fibrosis were measured by histology. Left ventricular expression of A- and B-type natriuretic peptides (ANP and BNP) were measured by qRT-PCR and circulating BNP level was measured by ELISA. In a subgroup of animals, hearts were perfused according to Langendorff and were subjected to 35 min global ischemia and 120 min reperfusion with or without IPRE (3 × 5 min I/R cycles applied before index ischemia). Then infarct size or phosphorylated and total forms of proteins related to the cardioprotective RISK (AKT, ERK1,2) and SAFE (STAT3) pathways were measured by Western blot. RESULTS The severity of CKD was similar in males and females. However, CKD males developed more severe LVH compared to females as assessed by echocardiography. Histology revealed cardiac fibrosis only in males in CKD. LV ANP expression was significantly increased due to CKD in both sexes, however, LV BNP and circulating BNP levels failed to significantly increase in CKD. In both sexes, IPRE significantly decreased the infarct size in both the sham-operated and CKD groups. IPRE significantly increased the phospho-STAT3/STAT3 ratio in sham-operated but not in CKD animals in both sexes. There were no significant differences in phospho-AKT/AKT and phospho-ERK1,2/ERK1,2 ratios between the groups. CONCLUSION The infarct size-limiting effect of IPRE was preserved in both sexes in CKD despite the more severe uremic cardiomyopathy in male CKD rats. Further research is needed to identify crucial molecular mechanisms in the cardioprotective effect of IPRE in CKD.
Collapse
|
40
|
O'Neill JD, Guenthart BA, Hozain AE, Bacchetta M. Xenogeneic support for the recovery of human donor organs. J Thorac Cardiovasc Surg 2021; 163:1563-1570. [PMID: 34607726 DOI: 10.1016/j.jtcvs.2021.07.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/16/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022]
Abstract
VIDEO ABSTRACT.
Collapse
Affiliation(s)
- John D O'Neill
- Department of Cell Biology, State University of New York Downstate Medical Center, SUNY Downstate Health Sciences University, Brooklyn, NY
| | | | - Ahmed E Hozain
- Department of Surgery, State University of New York Downstate Medical Center, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Matthew Bacchetta
- Department of Thoracic Surgery, Vanderbilt University, Nashville, Tenn; Department of Cardiac Surgery, Vanderbilt University, Nashville, Tenn; Department of Biomedical Engineering, Vanderbilt University, Nashville, Tenn.
| |
Collapse
|
41
|
Kaseda R, Hosojima M, Kuwahara S, Kabasawa H, Aoki H, Higuchi Y, Kon V, Narita I, Saito A. Rice Endosperm Protein Improves the Anti-Inflammatory Effects of High-Density Lipoprotein and Produces Lower Atherosclerotic Lesion Accelerated by the Renal Mass Reduction than Casein in a Mouse Model. J Am Coll Nutr 2021; 41:668-678. [PMID: 34424818 DOI: 10.1080/07315724.2021.1950584] [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: 10/20/2022]
Abstract
Chronic kidney disease (CKD) impairs the anti-inflammatory effects of high-density lipoprotein (HDL) and increases cardiovascular mortality. Though the potential role of dietary interventions to manage HDL is well studied, the clinical trials aimed to increase HDL levels have failed to reduce cardiovascular risk, rendering HDL function to be explored as a more relevant clinical parameter. This study investigates the effects of rice endosperm protein (REP), a plant-based protein, on the anti-inflammatory properties of HDL and renal injury-driven atherosclerosis in comparison with casein, an animal protein. Ten-week-old apolipoprotein E-deficient hyperlipidemic mice underwent uninephrectomy. The mice (n = 6 each) were pair-fed a normal casein-based diet or a REP-based diet (both with 20.0% protein content) for seven weeks. Atherosclerotic lesions were detected by en face Sudan IV staining of the aorta. The number and sizes of the atherosclerotic lesions were significantly lower in the REP-based diet-fed group than the casein-based diet-fed group (p = 0.038). However, the REP-based diet neither elicited an ameliorative effect on kidney function or histology nor impacted the cholesterol profiles. Furthermore, HDL from the REP-based diet-fed mice significantly suppressed the inflammatory cytokine response of human umbilical vein endothelial cells than that from the casein-based diet-fed mice (MCP-1, p = 0.010; IL-6, p = 0.011; IL-1β, p = 0.028). The REP-based diet has a higher potential to lessen the atherosclerotic lesions accelerated by renal mass reduction than a casein-based diet, which could be associated with the anti-inflammatory effects of HDL.
Collapse
Affiliation(s)
- Ryohei Kaseda
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Michihiro Hosojima
- Department of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Shoji Kuwahara
- Department of Applied Molecular Medicine, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Hideyuki Kabasawa
- Department of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Hiroyuki Aoki
- Department of Applied Molecular Medicine, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Yuki Higuchi
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan.,Rice Research Center, Kameda Seika Co. Ltd, Niigata, Japan
| | - Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennesse, USA
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Akihiko Saito
- Department of Applied Molecular Medicine, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| |
Collapse
|
42
|
Rosenkranz S. Pulmonale Hypertonie und Rechtsherzinsuffizienz. AKTUELLE KARDIOLOGIE 2021. [DOI: 10.1055/a-1511-2519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
ZusammenfassungPulmonale Hypertonie (PH) beschreibt einen hämodynamischen Zustand, der durch eine
Druckerhöhung im kleinen Kreislauf charakterisiert ist und der insbesondere bei älteren
Patienten häufig zu beobachten ist. In Abhängigkeit der Ätiologie differenziert die klinische
Klassifikation der pulmonalen Hypertonie 5 Hauptgruppen. Unabhängig von der zugrunde liegenden
Ursache führt eine PH zu einer chronischen Rechtsherzbelastung und Rechtsherzinsuffizienz.
Aktuelle Studien zeigen, dass bereits eine moderate Erhöhung des pulmonalarteriellen Druckes
sowie des pulmonalvaskulären Widerstands prognoserelevant sind. Im Einzelfall ist die
konsequente differenzialdiagnostische Abklärung einer PH essenziell, da sich die
Behandlungsoptionen je nach zugrunde liegender Ursache grundlegend unterscheiden.
Collapse
Affiliation(s)
- Stephan Rosenkranz
- Klinik für Kardiologie, Angiologie, Pneumologie und internistische Intensivmedizin, Universtiätsklinikum Köln, Köln, Deutschland
| |
Collapse
|
43
|
Singh S, Kanwar A, Sundaragiri PR, Cheungpasitporn W, Truesdell AG, Rab ST, Singh M, Vallabhajosyula S. Acute Kidney Injury in Cardiogenic Shock: An Updated Narrative Review. J Cardiovasc Dev Dis 2021; 8:88. [PMID: 34436230 PMCID: PMC8396972 DOI: 10.3390/jcdd8080088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/19/2022] Open
Abstract
Acute myocardial infarction with cardiogenic shock (AMI-CS) is associated with high mortality and morbidity despite advancements in cardiovascular care. AMI-CS is associated with multiorgan failure of non-cardiac organ systems. Acute kidney injury (AKI) is frequently seen in patients with AMI-CS and is associated with worse mortality and outcomes compared to those without. The pathogenesis of AMI-CS associated with AKI may involve more factors than previously understood. Early use of renal replacement therapies, management of comorbid conditions and judicious fluid administration may help improve outcomes. In this review, we seek to address the etiology, pathophysiology, management, and outcomes of AKI complicating AMI-CS.
Collapse
Affiliation(s)
- Sohrab Singh
- Department of Medicine, The Brooklyn Hospital, Brooklyn, NY 11201, USA;
| | - Ardaas Kanwar
- Department of Medicine, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA;
| | - Pranathi R. Sundaragiri
- Section of Primary Care Internal Medicine, Wake Forest Baptist Health, High Point, NC 27262, USA;
| | - Wisit Cheungpasitporn
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | | | - Syed Tanveer Rab
- Section of Interventional Cardiology, Division of Cardiovascular Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Mandeep Singh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Saraschandra Vallabhajosyula
- Section of Cardiovascular Medicine, Department of Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27262, USA
| |
Collapse
|
44
|
Prieto-Carrasco R, Silva-Palacios A, Rojas-Morales P, Aparicio-Trejo OE, Medina-Reyes EI, Hernández-Cruz EY, Sánchez-Garibay C, Salinas-Lara C, Pavón N, Roldán FJ, Zazueta C, Tapia E, Pedraza-Chaverri J. Unilateral Ureteral Obstruction for 28 Days in Rats Is Not Associated with Changes in Cardiac Function or Alterations in Mitochondrial Function. BIOLOGY 2021; 10:671. [PMID: 34356526 PMCID: PMC8301354 DOI: 10.3390/biology10070671] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/10/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022]
Abstract
Our work evaluated cardiac function and mitochondrial bioenergetics parameters in hearts from male Wistar rats subjected to the UUO model during 28 days of progression. We measured markers of kidney damage and inflammation in plasma and renal fibrosis by histological analysis and Western blot. Cardiac function was evaluated by echocardiography and proteins involved in cardiac damage by Western blot. Oxygen consumption and transmembrane potential were monitored in cardiac mitochondria using high-resolution respirometry. We also determined the activity of ATP synthase and antioxidant enzymes such as glutathione peroxidase, glutathione reductase, and catalase. Our results show that, although renal dysfunction is established in animals subjected to ureteral obstruction, cardiac function is maintained along with mitochondrial function and antioxidant enzymes activity after 28 days of injury evolution. Our results suggest that renocardiac syndrome might develop but belatedly in obstruction-induced renal damage, opening the opportunity for treatment to prevent this condition.
Collapse
Affiliation(s)
- Rodrigo Prieto-Carrasco
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (R.P.-C.); (P.R.-M.); (O.E.A.-T.); (E.I.M.-R.); (E.Y.H.-C.)
- Department of Cardio-Renal Pathophysiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico;
| | - Alejandro Silva-Palacios
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (A.S.-P.); (C.Z.)
| | - Pedro Rojas-Morales
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (R.P.-C.); (P.R.-M.); (O.E.A.-T.); (E.I.M.-R.); (E.Y.H.-C.)
- Department of Cardio-Renal Pathophysiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico;
| | - Omar Emiliano Aparicio-Trejo
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (R.P.-C.); (P.R.-M.); (O.E.A.-T.); (E.I.M.-R.); (E.Y.H.-C.)
| | - Estefany Ingrid Medina-Reyes
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (R.P.-C.); (P.R.-M.); (O.E.A.-T.); (E.I.M.-R.); (E.Y.H.-C.)
| | - Estefani Yaquelin Hernández-Cruz
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (R.P.-C.); (P.R.-M.); (O.E.A.-T.); (E.I.M.-R.); (E.Y.H.-C.)
| | - Carlos Sánchez-Garibay
- Department of Neuropathology, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (C.S.-G.); (C.S.-L.)
| | - Citlaltepetl Salinas-Lara
- Department of Neuropathology, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (C.S.-G.); (C.S.-L.)
| | - Natalia Pavón
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico;
| | - Francisco Javier Roldán
- Department of External Consultation, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico;
| | - Cecilia Zazueta
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (A.S.-P.); (C.Z.)
| | - Edilia Tapia
- Department of Cardio-Renal Pathophysiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico;
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (R.P.-C.); (P.R.-M.); (O.E.A.-T.); (E.I.M.-R.); (E.Y.H.-C.)
| |
Collapse
|
45
|
Yao M, Zhang C, Gao C, Wang Q, Dai M, Yue R, Sun W, Liang W, Zheng Z. Exploration of the Shared Gene Signatures and Molecular Mechanisms Between Systemic Lupus Erythematosus and Pulmonary Arterial Hypertension: Evidence From Transcriptome Data. Front Immunol 2021; 12:658341. [PMID: 34335565 PMCID: PMC8320323 DOI: 10.3389/fimmu.2021.658341] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/21/2021] [Indexed: 01/22/2023] Open
Abstract
Background Systemic lupus erythematosus (SLE) is an autoimmune disease that can affect multiple systems. Pulmonary arterial hypertension (PAH) has a close linkage with SLE. However, the inter-relational mechanisms between them are still unclear. This article aimed to explore the shared gene signatures and potential molecular mechanisms in SLE and PAH. Methods The microarray data of SLE and PAH in the Gene Expression Omnibus (GEO) database were downloaded. The Weighted Gene Co-Expression Network Analysis (WGCNA) was used to identify the co-expression modules related to SLE and PAH. The shared genes existing in the SLE and PAH were performed an enrichment analysis by ClueGO software, and their unique genes were also performed with biological processes analyses using the DAVID website. The results were validated in another cohort by differential gene analysis. Moreover, the common microRNAs (miRNAs) in SLE and PAH were obtained from the Human microRNA Disease Database (HMDD) and the target genes of whom were predicted through the miRTarbase. Finally, we constructed the common miRNAs–mRNAs network with the overlapped genes in target and shared genes. Results Using WGCNA, four modules and one module were identified as the significant modules with SLE and PAH, respectively. A ClueGO enrichment analysis of shared genes reported that highly activated type I IFN response was a common feature in the pathophysiology of SLE and PAH. The results of differential analysis in another cohort were extremely similar to them. We also proposed a disease road model for the possible mechanism of PAH secondary to SLE according to the shared and unique gene signatures in SLE and PAH. The miRNA–mRNA network showed that hsa-miR-146a might regulate the shared IFN-induced genes, which might play an important role in PAH secondary to SLE. Conclusion Our work firstly revealed the high IFN response in SLE patients might be a crucial susceptible factor for PAH and identified novel gene candidates that could be used as biomarkers or potential therapeutic targets.
Collapse
Affiliation(s)
- Menghui Yao
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunyi Zhang
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Congcong Gao
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qianqian Wang
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengmeng Dai
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Runzhi Yue
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenbo Sun
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenfang Liang
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhaohui Zheng
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
46
|
Bodine SC, Brooks HL, Bunnett NW, Coller HA, Frey MR, Joe B, Kleyman TR, Lindsey ML, Marette A, Morty RE, Ramírez JM, Thomsen MB, Yosten GLC. An American Physiological Society cross-journal Call for Papers on "Inter-Organ Communication in Homeostasis and Disease". Am J Physiol Lung Cell Mol Physiol 2021; 321:L42-L49. [PMID: 34010064 PMCID: PMC8321848 DOI: 10.1152/ajplung.00209.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 12/17/2022] Open
Affiliation(s)
- Sue C Bodine
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, New York University, New York, New York
| | - Hilary A Coller
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, California
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California
- Department of Biological Chemistry, University of California, Los Angeles, California
| | - Mark R Frey
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Bina Joe
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
- Center for Hypertension and Personalized Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Thomas R Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Hôpital Laval, Laval University, Quebec City, Québec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, Québec, Canada
| | - Rory E Morty
- Department of Translational Pulmonology and the Translational Lung Research Center Heidelberg, University Hospital Heidelberg, member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, Justus Liebig University Giessen, member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Jan-Marino Ramírez
- Department of Neurological Surgery, University of Washington Medical Center, Seattle, Washington
- Center on Human Development and Disability, University of Washington, Seattle, Washington
- Center for Integrative Brain Research at the Seattle Children's Research Institute, University of Washington, Seattle, Washington
| | - Morten B Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gina L C Yosten
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri
| |
Collapse
|
47
|
Conceptual advances and evolving terminology in acute kidney disease. Nat Rev Nephrol 2021; 17:493-502. [PMID: 33712812 DOI: 10.1038/s41581-021-00410-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 01/31/2023]
Abstract
Over the past decade, new insights into epidemiology, pathophysiology and biomarkers have modified our understanding of acute kidney dysfunction and damage, and their association with subsequent chronic kidney disease. The concept of acute kidney injury (AKI), which has relied on established but nonetheless flawed biomarkers of solute clearance (serum creatinine levels and urinary output), has been challenged by the identification of novel biomarkers of tubular stress and/or damage. The expression of some of these novel biomarkers precedes changes in conventional biomarkers or can increase their predictive power, and might therefore enhance the clinical accuracy of the definition of AKI. In addition, the need to consider AKI recurrence, duration and progression to chronic kidney disease within the clinical and epidemiological framework of AKI led to the emergence of the concept of acute kidney disease. New definitions of acute syndromes of kidney impairment and injury are needed.
Collapse
|
48
|
Possible benefits of exogenous melatonin for individuals on dialysis: a narrative review on potential mechanisms and clinical implications. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:1599-1611. [PMID: 34097094 DOI: 10.1007/s00210-021-02099-x] [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: 11/19/2020] [Accepted: 05/06/2021] [Indexed: 10/21/2022]
Abstract
Prevention of oxidative stress and inflammation in chronic kidney disease patients (CKD) on dialysis may reduce dialysis-associated complications. Administration of powerful antioxidants may improve the consequences of peritoneal dialysis (PD) and hemodialysis (HD). This narrative review aimed to show the potential therapeutic effects of melatonin (MLT) on the consequences of CKD patients receiving HD or PD. The results of preclinical and clinical studies have proven that CKD and dialysis are accompanied by reduced endogenous MLT levels and related complications such as sleep disorders. Enhanced oxidative stress, inflammation, cellular damages, and renal fibrosis, along with dysregulation of the renin-angiotensin system (RAS), have been observed in CKD and patients on dialysis. Results of studies have revealed that the restoration of MLT via the exogenous source may regulate oxidative stress, inflammation, and RAS functions, inhibit fibrosis, and improve complications in patients with long-term dialysis patients. In summary, treatment of patients with CKD and dialysis with exogenous MLT is suggested as a practical approach in reducing the outcomes and improving the quality of life in patients via antioxidant, anti-inflammatory, and anti-fibrotic signaling pathways. Therefore, this hormone can be considered in clinical practice to manage dialysis-related complications.
Collapse
|
49
|
Zhong J, Yang HC, Yermalitsky V, Shelton EL, Otsuka T, Wiese CB, May-Zhang LS, Banan B, Abumrad N, Huang J, Cavnar AB, Kirabo A, Yancey PG, Fogo AB, Vickers KC, Linton MF, Davies SS, Kon V. Kidney injury-mediated disruption of intestinal lymphatics involves dicarbonyl-modified lipoproteins. Kidney Int 2021; 100:585-596. [PMID: 34102217 DOI: 10.1016/j.kint.2021.05.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 04/06/2021] [Accepted: 05/06/2021] [Indexed: 12/24/2022]
Abstract
Kidney disease affects intestinal structure and function. Although intestinal lymphatics are central in absorption and remodeling of dietary and synthesized lipids/lipoproteins, little is known about how kidney injury impacts the intestinal lymphatic network, or lipoproteins transported therein. To study this, we used puromycin aminoglycoside-treated rats and NEP25 transgenic mice to show that proteinuric injury expanded the intestinal lymphatic network, activated lymphatic endothelial cells and increased mesenteric lymph flow. The lymph was found to contain increased levels of cytokines, immune cells, and isolevuglandin (a highly reactive dicarbonyl) and to have a greater output of apolipoprotein AI. Plasma levels of cytokines and isolevuglandin were not changed. However, isolevuglandin was also increased in the ileum of proteinuric animals, and intestinal epithelial cells exposed to myeloperoxidase produced more isolevuglandin. Apolipoprotein AI modified by isolevuglandin directly increased lymphatic vessel contractions, activated lymphatic endothelial cells, and enhanced the secretion of the lymphangiogenic promoter vascular endothelial growth factor-C by macrophages. Inhibition of isolevuglandin synthesis by a carbonyl scavenger reduced intestinal isolevuglandin adduct level and lymphangiogenesis. Thus, our data reveal a novel mediator, isolevuglandin modified apolipoprotein AI, and uncover intestinal lymphatic network structure and activity as a new pathway in the crosstalk between kidney and intestine that may contribute to the adverse impact of kidney disease on other organs.
Collapse
Affiliation(s)
- Jianyong Zhong
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hai-Chun Yang
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| | - Valery Yermalitsky
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Elaine L Shelton
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tadashi Otsuka
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Carrie B Wiese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Linda S May-Zhang
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Babak Banan
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Naji Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jiansheng Huang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ashley B Cavnar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annet Kirabo
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Patricia G Yancey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Agnes B Fogo
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kasey C Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - MacRae F Linton
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sean S Davies
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
50
|
Hospitalization for acute coronary syndrome increases the long-term risk of pneumonia: a population-based cohort study. Sci Rep 2021; 11:9696. [PMID: 33958673 PMCID: PMC8102567 DOI: 10.1038/s41598-021-89038-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/15/2021] [Indexed: 11/30/2022] Open
Abstract
It is well established that the risk of acute coronary syndrome (ACS) increases after respiratory infection. However, the reverse association has not been evaluated. We tested the hypothesis that the long-term risk of pneumonia is increased after a new ACS event. A matched-cohort study was conducted using a nationally representative dataset. We identified patients with admission for ACS between 2004 and 2014, without a previous history of ACS or pneumonia. Incidence density sampling was used to match patients, on the basis of age and sex, to 3 controls who were also free from both ACS and pneumonia. We examined the incidence of pneumonia after ACS until the end of the cohort observation (Dec 31, 2014). The analysis cohort consisted of 5469 ACS cases and 16,392 controls (median age, 64 years; 68.3% men). The incidence rate ratios of the first and the total pneumonia episodes in the ACS group relative to the control group was 1.25 (95% confidence interval [CI], 1.11–1.41) and 1.23(95% CI 1.11–1.36), respectively. A significant ACS-related increase in the incidence of pneumonia was observed in the Cox-regression, shared frailty, and joint frailty model analyses, with hazard ratios of 1.25 (95% CI 1.09–1.42), 1.35 (95% CI 1.15–1.58), and 1.24 (95% CI 1.10–1.39), respectively. In this population-based cohort of patients who were initially free from both ACS and pneumonia, we found that hospitalization for ACS substantially increased the long term risk of pneumonia. This should be considered when formulating post-discharge care plans and preventive vaccination strategies in patients with ACS.
Collapse
|