1
|
Renberg M, Karlsson T, Gellerfors M, Gustavsson J, Wellfelt K, Günther M. Intramuscular vasopressin: A feasible intervention for prehospital hemodynamic stabilization in porcine hemorrhagic shock and whole blood transfusion. Transfusion 2025; 65 Suppl 1:S68-S79. [PMID: 40123085 PMCID: PMC12035989 DOI: 10.1111/trf.18218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Hemorrhagic shock is the leading cause of preventable prehospital trauma deaths. While arginine vasopressin (AVP) is a well-known hormone with vasopressor effects, its potential for hemodynamic stabilization in prehospital hemorrhagic shock remains underexplored. This study investigated intramuscular (IM) AVP during hemorrhagic shock to evaluate its feasibility and efficacy for prehospital trauma resuscitation. STUDY DESIGN AND METHODS In this randomized, controlled, double-blinded trial, 16 swine (mean [standard deviation, SD] weight 56.2 [3.8] kg) underwent a mean (SD) 1205 (124) mL Class III hemorrhage for 45 min and 45 min of hypotension. Animals were randomized to 40 U IM AVP (n = 7) or NaCl (n = 9), followed immediately by 500 mL autologous whole blood transfusion over 30 and 120 min posttransfusion monitoring of hemodynamic, respiratory, and metabolic parameters. RESULTS AVP increased systolic arterial pressure 30 min after administration (mean increase: 33.5 mmHg vs. 7.5 mmHg, p < 0.05) and improved cardiac index (CI) 90 min after AVP (mean increase: 19.2% vs. 4.1% decrease, p < 0.05) and stroke volume (mean increase: 37.0% vs. 1.0%, p < 0.05). These effects normalized by 120 min. AVP did not affect respiratory parameters, oxygen delivery, or consumption. Increased serum AVP confirmed systemic uptake (median 68.7 pg/mL vs. 10.0 pg/mL in controls, p < 0.05). CONCLUSION IM AVP, combined with whole blood transfusion, transiently stabilized hemodynamics by increasing systemic vascular resistance index, systolic blood pressure, and CI without respiratory compromise. These findings suggest that IM AVP may be a viable intervention for prehospital resuscitation of severe hemorrhagic shock, offering vital short-term stabilization to facilitate transport to definitive care.
Collapse
Affiliation(s)
- Mattias Renberg
- Department of Clinical Science and Education, SödersjukhusetSection for Anesthesiology and Intensive Care, Karolinska InstitutetStockholmSweden
| | - Tomas Karlsson
- Department of Clinical Science and Education, SödersjukhusetSection for Anesthesiology and Intensive Care, Karolinska InstitutetStockholmSweden
- Rapid Response Car AISABStockholmSweden
| | - Mikael Gellerfors
- Department of Physiology and Pharmacology, Section of Anesthesiology and Intensive CareKarolinska InstitutetStockholmSweden
- Department of Perioperative Medicine and Intensive CareKarolinska University HospitalStockholmSweden
- Swedish Air Ambulance (SLA)MoraSweden
- Ambulance Helicopter and Rapid Response Car AISABStockholmSweden
| | | | - Katrin Wellfelt
- Department of NeuroscienceKarolinska InstitutetStockholmSweden
| | - Mattias Günther
- Department of Clinical Science and Education, SödersjukhusetSection for Anesthesiology and Intensive Care, Karolinska InstitutetStockholmSweden
- Department of NeuroscienceKarolinska InstitutetStockholmSweden
| |
Collapse
|
2
|
Bezati S, Ventoulis I, Bistola V, Verras C, Matsiras D, Polyzogopoulou E, Parissis J. Copeptin in Acute Myocardial Infarction: Is There a Role in the Era of High-Sensitivity Troponins? J Cardiovasc Dev Dis 2025; 12:144. [PMID: 40278203 PMCID: PMC12027642 DOI: 10.3390/jcdd12040144] [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: 02/19/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025] Open
Abstract
The quest for prompt and effective diagnosis of acute myocardial infarction (AMI) has been in the spotlight for decades. Ongoing research focuses on refined biomarker strategies for the early identification and disposition of patients with symptoms suggestive of AMI. Copeptin, a surrogate of the hormone arginine vasopressin, has emerged as a novel biomarker that could potentially aid in the diagnostic approach of patients with chest pain presenting to the emergency department. Observational studies have demonstrated that copeptin is upregulated in patients with AMI, although the exact pathophysiological mechanisms implicated in its release during myocardial ischemia remain unclear. Following these observations, copeptin was proposed as an adjunct to troponin in an effort to augment the diagnostic accuracy of conventional troponin assays. However, after the introduction of high-sensitivity troponin assays, the diagnostic utility of copeptin has been debated. This narrative review aims to elucidate plausible pathophysiological mechanisms involved in copeptin release during myocardial ischemia and to summarize the most recent evidence regarding its diagnostic potential in combination with high-sensitivity troponin assays.
Collapse
Affiliation(s)
- Sofia Bezati
- Department of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.B.); (C.V.); (D.M.); (J.P.)
| | - Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, 50200 Ptolemaida, Greece;
| | - Vasiliki Bistola
- Second Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Christos Verras
- Department of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.B.); (C.V.); (D.M.); (J.P.)
| | - Dionysis Matsiras
- Department of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.B.); (C.V.); (D.M.); (J.P.)
| | - Effie Polyzogopoulou
- Department of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.B.); (C.V.); (D.M.); (J.P.)
| | - John Parissis
- Department of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.B.); (C.V.); (D.M.); (J.P.)
| |
Collapse
|
3
|
Kato H, Mathis BJ, Shimoda T, Nakajima T, Tokunaga C, Hiramatsu Y. Hemodynamic Management with Vasopressin for Cardiovascular Surgery. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:2064. [PMID: 39768943 PMCID: PMC11676985 DOI: 10.3390/medicina60122064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/21/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: Vasopressin increases blood pressure through aquaporin-2-mediated water retention and is useful for managing hemodynamics after surgery. However, even after decades of study, clear clinical guidelines on doses and ideal use cases after cardiovascular surgery remain unclear. Here, the existing literature is synthesized on vasopressin use for cardiac surgeries and coupled with real-world clinical experience to outline a clearer clinical path for vasopressin use. Materials and Methods: Literature from 1966 to the present was searched, and information on surgical outcomes for cardiovascular surgery was extracted. Clinicians from the University of Tsukuba with extensive vasopressin experience in pediatric cardiovascular patients were consulted for general use guidelines. Results: Vasopressin response after cardiovascular surgery is multifaceted, and low-power trials, plus conflicting study reports, generally render it as a secondary choice behind norepinephrine. Clinical experience indicates that low doses of 0.2-0.3 mU/kg/min with constant blood pressure and oxygen monitoring for response are required. Although sole use is not recommended, vasopressin may aid in controlling hemodynamics when given with other volemic or osmolal drugs. Conclusions: Vasopressin may work in a select population of first-line non-responders, but relevant response factors remain unanalyzed and clear guidelines for use remain unestablished. Future, large-scale studies are needed to delineate temporal and demographic characteristics that affect response to vasopressin for the purpose of managing post-surgical capillary leakage and hemodynamics.
Collapse
|
4
|
Szczepanska-Sadowska E, Czarzasta K, Bogacki-Rychlik W, Kowara M. The Interaction of Vasopressin with Hormones of the Hypothalamo-Pituitary-Adrenal Axis: The Significance for Therapeutic Strategies in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2024; 25:7394. [PMID: 39000501 PMCID: PMC11242374 DOI: 10.3390/ijms25137394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
A large body of evidence indicates that vasopressin (AVP) and steroid hormones are frequently secreted together and closely cooperate in the regulation of blood pressure, metabolism, water-electrolyte balance, and behavior, thereby securing survival and the comfort of life. Vasopressin cooperates with hormones of the hypothalamo-pituitary-adrenal axis (HPA) at several levels through regulation of the release of corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), and multiple steroid hormones, as well as through interactions with steroids in the target organs. These interactions are facilitated by positive and negative feedback between specific components of the HPA. Altogether, AVP and the HPA cooperate closely as a coordinated functional AVP-HPA system. It has been shown that cooperation between AVP and steroid hormones may be affected by cellular stress combined with hypoxia, and by metabolic, cardiovascular, and respiratory disorders; neurogenic stress; and inflammation. Growing evidence indicates that central and peripheral interactions between AVP and steroid hormones are reprogrammed in cardiovascular and metabolic diseases and that these rearrangements exert either beneficial or harmful effects. The present review highlights specific mechanisms of the interactions between AVP and steroids at cellular and systemic levels and analyses the consequences of the inappropriate cooperation of various components of the AVP-HPA system for the pathogenesis of cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | | | | | | |
Collapse
|
5
|
De Simone B, Chouillard E, Podda M, Pararas N, de Carvalho Duarte G, Fugazzola P, Birindelli A, Coccolini F, Polistena A, Sibilla MG, Kruger V, Fraga GP, Montori G, Russo E, Pintar T, Ansaloni L, Avenia N, Di Saverio S, Leppäniemi A, Lauretta A, Sartelli M, Puzziello A, Carcoforo P, Agnoletti V, Bissoni L, Isik A, Kluger Y, Moore EE, Romeo OM, Abu-Zidan FM, Beka SG, Weber DG, Tan ECTH, Paolillo C, Cui Y, Kim F, Picetti E, Di Carlo I, Toro A, Sganga G, Sganga F, Testini M, Di Meo G, Kirkpatrick AW, Marzi I, déAngelis N, Kelly MD, Wani I, Sakakushev B, Bala M, Bonavina L, Galante JM, Shelat VG, Cobianchi L, Mas FD, Pikoulis M, Damaskos D, Coimbra R, Dhesi J, Hoffman MR, Stahel PF, Maier RV, Litvin A, Latifi R, Biffl WL, Catena F. The 2023 WSES guidelines on the management of trauma in elderly and frail patients. World J Emerg Surg 2024; 19:18. [PMID: 38816766 PMCID: PMC11140935 DOI: 10.1186/s13017-024-00537-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/26/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND The trauma mortality rate is higher in the elderly compared with younger patients. Ageing is associated with physiological changes in multiple systems and correlated with frailty. Frailty is a risk factor for mortality in elderly trauma patients. We aim to provide evidence-based guidelines for the management of geriatric trauma patients to improve it and reduce futile procedures. METHODS Six working groups of expert acute care and trauma surgeons reviewed extensively the literature according to the topic and the PICO question assigned. Statements and recommendations were assessed according to the GRADE methodology and approved by a consensus of experts in the field at the 10th international congress of the WSES in 2023. RESULTS The management of elderly trauma patients requires knowledge of ageing physiology, a focused triage, including drug history, frailty assessment, nutritional status, and early activation of trauma protocol to improve outcomes. Acute trauma pain in the elderly has to be managed in a multimodal analgesic approach, to avoid side effects of opioid use. Antibiotic prophylaxis is recommended in penetrating (abdominal, thoracic) trauma, in severely burned and in open fractures elderly patients to decrease septic complications. Antibiotics are not recommended in blunt trauma in the absence of signs of sepsis and septic shock. Venous thromboembolism prophylaxis with LMWH or UFH should be administrated as soon as possible in high and moderate-risk elderly trauma patients according to the renal function, weight of the patient and bleeding risk. A palliative care team should be involved as soon as possible to discuss the end of life in a multidisciplinary approach considering the patient's directives, family feelings and representatives' desires, and all decisions should be shared. CONCLUSIONS The management of elderly trauma patients requires knowledge of ageing physiology, a focused triage based on assessing frailty and early activation of trauma protocol to improve outcomes. Geriatric Intensive Care Units are needed to care for elderly and frail trauma patients in a multidisciplinary approach to decrease mortality and improve outcomes.
Collapse
Affiliation(s)
- Belinda De Simone
- Department of Emergency Minimally Invasive Surgery, Academic Hospital of Villeneuve St Georges, Villeneuve St Georges, France.
- Department of General Minimally Invasive Surgery, Infermi Hospital, AUSL Romagna, Rimini, Italy.
- General Surgery Department, American Hospital of Paris, Paris, France.
| | - Elie Chouillard
- General Surgery Department, American Hospital of Paris, Paris, France
| | - Mauro Podda
- Department of Surgical Science, Unit of Emergency Surgery, University of Cagliari, Cagliari, Italy
| | - Nikolaos Pararas
- 3rd Department of Surgery, Attikon General Hospital, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | | | - Paola Fugazzola
- Unit of General Surgery I, IRCCS San Matteo Hospital of Pavia, University of Pavia, Pavia, Italy
| | | | | | - Andrea Polistena
- Department of Surgery, Policlinico Umberto I Roma, Sapienza University, Rome, Italy
| | - Maria Grazia Sibilla
- Department of Surgery, Unit of General Surgery, University Hospital of Ferrara and University of Ferrara, Ferrara, Italy
| | - Vitor Kruger
- Division of Trauma Surgery, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Gustavo P Fraga
- Division of Trauma Surgery, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Giulia Montori
- Unit of General and Emergency Surgery, Vittorio Veneto Hospital, Via C. Forlanini 71, 31029, Vittorio Veneto, TV, Italy
| | - Emanuele Russo
- Department of Anesthesia, Level I, Trauma Center, Bufalini Hospital, Cesena, Italy
| | - Tadeja Pintar
- UMC Ljubljana and Medical Faculty Ljubljana, Ljubljana, Slovenia
| | - Luca Ansaloni
- New Zealand Blood Service, Christchurch, New Zealand
| | - Nicola Avenia
- Endocrine Surgical Unit - University of Perugia, Terni, Italy
| | - Salomone Di Saverio
- General Surgery Unit, Madonna del Soccorso Hospital, AST Ascoli Piceno, San Benedetto del Tronto, Italy
| | - Ari Leppäniemi
- Division of Emergency Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Andrea Lauretta
- Department of Surgical Oncology, Centro Di Riferimento Oncologico Di Aviano IRCCS, Aviano, Italy
| | - Massimo Sartelli
- Department of General Surgery, Macerata Hospital, Macerata, Italy
| | - Alessandro Puzziello
- Dipartimento di Medicina, Chirurgia e Odontoiatria, Campus Universitario di Baronissi (SA) - Università di Salerno, AOU San Giovanni di Dio e Ruggi di Aragona, Salerno, Italy
| | - Paolo Carcoforo
- Department of Surgery, Unit of General Surgery, University Hospital of Ferrara and University of Ferrara, Ferrara, Italy
| | - Vanni Agnoletti
- Department of Anesthesia, Level I, Trauma Center, Bufalini Hospital, Cesena, Italy
| | - Luca Bissoni
- Department of Anesthesia, Level I, Trauma Center, Bufalini Hospital, Cesena, Italy
| | - Arda Isik
- Istanbul Medeniyet University, Istanbul, Turkey
| | - Yoram Kluger
- Department of General Surgery, Rambam Health Care Campus, Haifa, Israel
| | - Ernest E Moore
- Ernest E Moore Shock Trauma Center at Denver Health, University of Colorado, Denver, CO, USA
| | - Oreste Marco Romeo
- Bronson Methodist Hospital/Western Michigan University, Kalamazoo, MI, USA
| | - Fikri M Abu-Zidan
- Department of Surgery, College of Medicine and Health Sciences, United Arab Emirates University, Al‑Ain, United Arab Emirates
| | | | - Dieter G Weber
- Department of General Surgery, Royal Perth Hospital and The University of Western Australia, Perth, Australia
| | - Edward C T H Tan
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ciro Paolillo
- Emergency Department, Ospedale Civile Maggiore, Verona, Italy
| | - Yunfeng Cui
- Department of Surgery, Tianjin Nankai Hospital, Nankai Clinical School of Medicine, Tianjin Medical University, Tianjin, China
| | - Fernando Kim
- University of Colorado Anschutz Medical Campus, Denver, CO, 80246, USA
| | - Edoardo Picetti
- Department of Anesthesia and Intensive Care, Parma University Hospital, Parma, Italy
| | - Isidoro Di Carlo
- Department of Surgical Sciences and Advanced Technologies, General Surgery Cannizzaro Hospital, University of Catania, Catania, Italy
| | - Adriana Toro
- Department of Surgical Sciences and Advanced Technologies, General Surgery Cannizzaro Hospital, University of Catania, Catania, Italy
| | - Gabriele Sganga
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University, Rome, Italy
| | - Federica Sganga
- Department of Geriatrics, Ospedale Sant'Anna, Ferrara, Italy
| | - Mario Testini
- Department of Precision and Regenerative Medicine and Ionian Area, Unit of Academic General Surgery, University of Bari "A. Moro", Bari, Italy
| | - Giovanna Di Meo
- Department of Precision and Regenerative Medicine and Ionian Area, Unit of Academic General Surgery, University of Bari "A. Moro", Bari, Italy
| | - Andrew W Kirkpatrick
- Departments of Surgery and Critical Care Medicine, University of Calgary, Foothills Medical Centre, Calgary, AB, Canada
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Frankfurt, Germany
| | - Nicola déAngelis
- Unit of Colorectal and Digestive Surgery, DIGEST Department, Beaujon University Hospital, AP-HP, University of Paris Cité, Clichy, France
| | | | - Imtiaz Wani
- Department of Surgery, Government Gousia Hospital, DHS, Srinagar, India
| | - Boris Sakakushev
- General Surgery Department, Medical University, University Hospital St George, Plovdiv, Bulgaria
| | - Miklosh Bala
- Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Luigi Bonavina
- Division of General Surgery, IRCCS Policlinico San Donato, University of Milan, Milan, Italy
| | - Joseph M Galante
- Division of Trauma and Acute Care Surgery, Department of Surgery, University of California Davis, Sacramento, CA, USA
| | - Vishal G Shelat
- Department of General Surgery, Tan Tock Seng Hospital, Novena, Singapore
| | - Lorenzo Cobianchi
- Unit of General Surgery I, IRCCS San Matteo Hospital of Pavia, University of Pavia, Pavia, Italy
- Collegium Medicum, University of Social Sciences, Łodz, Poland
| | - Francesca Dal Mas
- Department of Management, Ca' Foscari University of Venice, Venice, Italy
- Collegium Medicum, University of Social Sciences, Łodz, Poland
| | - Manos Pikoulis
- Department of Surgical Science, Unit of Emergency Surgery, University of Cagliari, Cagliari, Italy
| | | | - Raul Coimbra
- Riverside University Health System Medical Center, Riverside, CA, USA
| | - Jugdeep Dhesi
- Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Melissa Red Hoffman
- Department of Surgery, University of North Carolina, Surgical Palliative Care Society, Asheville, NC, USA
| | - Philip F Stahel
- Department of Surgery, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Ronald V Maier
- Harborview Medical Center, University of Washington, Seattle, WA, USA
| | - Andrey Litvin
- Department of Surgical Diseases No. 3, Gomel State Medical University, University Clinic, Gomel, Belarus
| | - Rifat Latifi
- University of Arizona, Tucson, AZ, USA
- Abrazo Health West Campus, Goodyear, Tucson, AZ, USA
| | - Walter L Biffl
- Division of Trauma/Acute Care Surgery, Scripps Clinic Medical Group, La Jolla, CA, USA
| | - Fausto Catena
- Department of General and Emergency Surgery, Bufalini Hospital-Level 1 Trauma Center, AUSL Romagna, Cesena, Italy
| |
Collapse
|
6
|
Hwang KY, Phoon PHY, Hwang NC. Adverse Clinical Effects Associated With Non-catecholamine Pharmacologic Agents for Treatment of Vasoplegic Syndrome in Adult Cardiac Surgery. J Cardiothorac Vasc Anesth 2024; 38:802-819. [PMID: 38218651 DOI: 10.1053/j.jvca.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/23/2023] [Accepted: 12/11/2023] [Indexed: 01/15/2024]
Abstract
Vasoplegic syndrome is a relatively common complication that can happen during and after major adult cardiac surgery. It is associated with a higher rate of complications, including postoperative renal failure, longer duration of mechanical ventilation, and intensive care unit stay, as well as increased mortality. The underlying pathophysiology of vasoplegic syndrome is that of profound vascular hyporesponsiveness, and involves a complex interplay among inflammatory cytokines, cellular surface receptors, and nitric oxide (NO) production. The pharmacotherapy approaches for the treatment of vasoplegia include medications that increase vascular smooth muscle contraction via increasing cytosolic calcium in myocytes, reduce the vascular effects of NO and inflammation, and increase the biosynthesis of and vascular response to norepinephrine. Clinical trials have demonstrated the clinical efficacy of non-catecholamine pharmacologic agents in the treatment of vasoplegic syndrome. With an increase in their use today, it is important for clinicians to understand the adverse clinical outcomes and patient risk profiles associated with these agents, which will allow better-tailored medical therapy.
Collapse
Affiliation(s)
- Kai Yin Hwang
- Department of Anaesthesiology, National University Hospital, Singapore
| | - Priscilla Hui Yi Phoon
- Department of Anaesthesiology, Singapore General Hospital, Singapore; Department of Cardiothoracic Anaesthesia, National Heart Centre, Singapore
| | - Nian Chih Hwang
- Department of Anaesthesiology, Singapore General Hospital, Singapore; Department of Cardiothoracic Anaesthesia, National Heart Centre, Singapore.
| |
Collapse
|
7
|
Trieu NHK, Pham HM, Mai AT. Initial management of acute circulatory failure in amniotic fluid embolism: A narrative review. TRENDS IN ANAESTHESIA AND CRITICAL CARE 2023; 52:101288. [DOI: 10.1016/j.tacc.2023.101288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
|
8
|
Bulloch MN. Letter to the editor: "Combination of norepinephrine with phenylephrine versus norepinephrine with vasopressin in critically ill patients with septic shock: A retrospective study". J Crit Care 2023; 74:154239. [PMID: 36549962 DOI: 10.1016/j.jcrc.2022.154239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Marilyn N Bulloch
- Department of Pharmacy Practice, Auburn University Harrison College of Pharmacy, United States of America; Department of Family, Internal, and Rural Medicine College of Community Health Sciences, University of Alabama School of Medicine, Northeast Medical Building, Box 3611, Tuscaloosa, AL 35091, United States of America.
| |
Collapse
|
9
|
Capolupo I, De Rose DU, Mazzeo F, Monaco F, Giliberti P, Landolfo F, Di Pede A, Toscano A, Conforti A, Bagolan P, Dotta A. Early vasopressin infusion improves oxygenation in infants with congenital diaphragmatic hernia. Front Pediatr 2023; 11:1104728. [PMID: 37063685 PMCID: PMC10090559 DOI: 10.3389/fped.2023.1104728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/14/2023] [Indexed: 04/18/2023] Open
Abstract
Objective Congenital Diaphragmatic Hernia (CDH) is a complex disease including a diaphragmatic defect, lung hypoplasia, and pulmonary hypertension. Despite its increasing use in neonates, the literature on the use of vasopressin in neonates is limited. The aim of this work is to analyze the changes in clinical and hemodynamic variables in a cohort of CDH infants treated with vasopressin. Methods Among CDH infants managed at the Neonatal Intensive Care Unit (NICU) of our hospital from May 2014 to January 2019, all infants who were treated with vasopressin, because of systemic hypotension and pulmonary hypertension, were enrolled in this retrospective study. The primary outcome was the change in oxygenation index (OI) after the start of the infusion of vasopressin. The secondary outcomes were the changes in cerebral and splanchnic fractional tissue oxygen extraction (FTOEc and FTOEs) at near-infrared spectroscopy, to understand the balance between oxygen supply and tissue oxygen consumption after the start of vasopressin infusion. We also reported as secondary outcomes the changes in ratio of arterial oxygen partial pressure (PaO2) to fraction of inspired oxygen (FiO2), heart rate, mean arterial pressure, serum pH, and serum sodium. Results We included 27 patients with isolated CDH who received vasopressin administration. OI dramatically dropped when vasopressin infusion started, with a significant reduction according to ANOVA for repeated measures (p = 0.003). A global significant improvement in FTOEc and FTOEs was detected (p = 0.009 and p = 0.004, respectively) as a significant reduction in heart rate (p = 0.019). A global significant improvement in PaO2/FiO2 ratio was observed (p < 0.001) and also at all time points: at 6 h since infusion (p = 0.015), 12 h (p = 0.009), and 24 h (p = 0.006), respectively. A significant reduction in sodium levels was observed as expected side effect (p = 0.012). No significant changes were observed in the remaining outcomes. Conclusion Our data suggest that starting early vasopressin infusion in CDH infants with pulmonary hypertension could improve oxygenation index and near-infrared spectroscopy after 12 and 24 h of infusion. These pilot data represent a background for planning future larger randomized trials to evaluate the efficacy and safety of vasopressin for the CDH population.
Collapse
Affiliation(s)
- Irma Capolupo
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus – Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Domenico Umberto De Rose
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus – Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Francesca Mazzeo
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus – Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Francesca Monaco
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus – Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Paola Giliberti
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus – Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Francesca Landolfo
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus – Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Alessandra Di Pede
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus – Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Alessandra Toscano
- Perinatal Cardiology, Medical and Surgical Department of Fetus – Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Andrea Conforti
- Neonatal Surgery Unit, Medical and Surgical Department of Fetus – Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Pietro Bagolan
- Neonatal Surgery Unit, Medical and Surgical Department of Fetus – Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
- Department of Systems Medicine, University of Tor Vergata, Rome, Italy
| | - Andrea Dotta
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus – Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| |
Collapse
|
10
|
Barańska-Pawełczak K, Wojciechowska C, Jacheć W. Pregnancy in Patients with Pulmonary Arterial Hypertension in Light of New ESC Guidelines on Pulmonary Hypertension. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4625. [PMID: 36901635 PMCID: PMC10001459 DOI: 10.3390/ijerph20054625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 06/18/2023]
Abstract
Pulmonary arterial hypertension (PAH) is defined as an elevated mean pulmonary artery pressure (mPAP) of >20 mmHg together with a pulmonary arterial wedge pressure (PAWP) of ≤15 mmHg and pulmonary vascular resistance (PVR) of>2 Wood units (WU). Although the total mortality of pregnant women with PAH has decreased significantly in recent years and is reported to be around 12% in some databases, total mortality is still at an unacceptably high percentage. Moreover, some subgroups, such as patients with Eisenmenger's syndrome, have a particularly high mortality rate of up to 36%. Pregnancy in patients with PAH is contraindicated; its appearance is an indication for a planned termination. Education of patients with PAH, including counseling on effective contraception, is essential. During pregnancy, blood volume, heart rate, and cardiac output increase, while PVR and systemic vascular resistance decrease. The hemostatic balance is shifted towards hypercoagulability. Among PAH-specific drugs, the use of inhaled or intravenous prostacyclins, phosphodiesterase inhibitors, and calcium channel blockers (in patients with preserved vasoreactivity) is acceptable. Endothelin receptor antagonists and riociguat are contraindicated. Childbirth can take place through either vaginal delivery or caesarean section; similarly, neuraxial and general anesthesia have proven indications. In a situation where all pharmacological options have been used in pregnant or postpartum patients in a serious condition, veno-arterial ECMO is a useful therapeutic option. For PAH patients who want to become mothers, an option that does not endanger their lives is adoption.
Collapse
Affiliation(s)
| | - Celina Wojciechowska
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| | - Wojciech Jacheć
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
11
|
Al-Saadi A, Sushko K, Bui V, van den Anker J, Razak A, Samiee-Zafarghandy S. Efficacy and Safety of Vasopressin and Terlipressin in Preterm Neonates: A Systematic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:13760. [PMID: 36360641 PMCID: PMC9658127 DOI: 10.3390/ijerph192113760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 06/16/2023]
Abstract
INTRODUCTION The use of arginine vasopressin (AVP) and terlipressin to treat hypotension in preterm neonates is increasing. Our aim was to review the available evidence on the efficacy and safety of AVP and terlipressin for use in preterm neonates. METHODS MEDLINE, EMBASE, the Cochrane Central Register of Controlled Trials, Web of Science, and Google Scholar from inception to September 2021 were searched for studies of AVP and terlipressin in the treatment of hypotension of any cause in preterm neonates. Primary outcomes were improvement in end-organ perfusion and mortality. The risk of bias assessment and certainty of the evidence were performed using appropriate tools. RESULTS Fifteen studies describing the use of AVP (n = 12) or terlipressin (n = 3) among 148 preterm neonates were included. Certainly, the available evidence for the primary outcome of end-organ perfusion rated as very low. AVP or terlipressin were used to treat 144 and 4 neonates, respectively. Improvement in markers of end-organ perfusion was reported in 143 (99%) neonates treated with AVP and 3 (75%) treated with terlipressin. The mortality rate was 41% (n = 59) and 50% (n = 2) for neonates who received AVP and terlipressin, respectively. Hyponatremia was the most frequently reported adverse event (n = 37, 25%). CONCLUSION AVP and terlipressin may improve measured blood pressure values and possibly end-organ perfusion among neonates with refractory hypotension. However, the efficacy-safety balance of these drugs should be assessed on an individual basis and as per the underlying cause. Studies on the optimal dosing, efficacy, and safety of AVP and terlipressin in preterm neonates with variable underlying conditions are critically needed.
Collapse
Affiliation(s)
- Abdulrahman Al-Saadi
- Division of Neonatology, Department of Pediatrics, Sultan Qaboos University, Muscat 123, Oman
| | - Katelyn Sushko
- Faculty of Health Sciences, School of Nursing, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Vivian Bui
- Department of Pharmacy, Hamilton Health Sciences, Hamilton, ON L8L 2X2, Canada
| | - John van den Anker
- Pediatric Pharmacology and Pharmacometrics, University Children’s Hospital Basel (UKBB), University of Basel, 4055 Basel, Switzerland
- Division of Clinical Pharmacology, Children’s National Hospital, Washington, DC 20010, USA
- Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children’s Hospital, 3000 CB Rotterdam, The Netherlands
| | - Abdul Razak
- Division of Neonatology, Department of Pediatrics, King Abdullah bin Abdulaziz University Hospital, Princess Norah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia
- Department of Pediatrics, Monash University, Melbourne 3800, Australia
| | - Samira Samiee-Zafarghandy
- Division of Neonatology, Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
12
|
Ferlini L, Nonclercq A, Su F, Creteur J, Taccone FS, Gaspard N. Sepsis modulates cortical excitability and alters the local and systemic hemodynamic response to seizures. Sci Rep 2022; 12:11336. [PMID: 35790848 PMCID: PMC9256588 DOI: 10.1038/s41598-022-15426-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/23/2022] [Indexed: 11/09/2022] Open
Abstract
Non-convulsive seizures and status epilepticus are frequent and associated with increased mortality in septic patients. However, the mechanism through which seizures impact outcome in these patients is unclear. As previous studies yielded an alteration of neurovascular coupling (NVC) during sepsis, we hypothesized that non-convulsive seizures, might further impair NVC, leading to brain tissue hypoxia. We used a previously developed ovine model of sepsis. Animals were allocated to sham procedure or sepsis; septic animals were studied either during the hyperdynamic phase (sepsis group) or after septic shock occurrence (septic shock group). After allocation, seizures were induced by cortical application of penicillin. We recorded a greater seizure-induced increase in the EEG gamma power in the sepsis group than in sham. Using a neural mass model, we also found that the theoretical activity of the modeled inhibitory interneurons, thought to be important to reproduce gamma oscillations, were relatively greater in the sepsis group. However, the NVC was impaired in sepsis animals, despite a normal brain tissue oxygenation. In septic shock animals, it was not possible to induce seizures. Cortical activity declined in case of septic shock, but it did not differ between sham or sepsis animals. As the alteration in NVC preceded cortical activity reduction, we suggest that, during sepsis progression, the NVC inefficiency could be partially responsible for the alteration of brain function, which might prevent seizure occurrence during septic shock. Moreover, we showed that cardiac output decreased during seizures in sepsis animals instead of increasing as in shams. The alteration of the seizure-induced systemic hemodynamic variations in sepsis might further affect cerebrovascular response to neuronal activation. Our findings support the hypothesis that anomalies in the cerebral blood flow regulation may contribute to the sepsis-associated encephalopathy and that seizures might be dangerous in such a vulnerable setting.
Collapse
Affiliation(s)
- Lorenzo Ferlini
- Department of Neurology, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik, 808, 1070, Brussels, Belgium
| | - Antoine Nonclercq
- Bio-, Electro- And Mechanical Systems (BEAMS), Université Libre de Bruxelles, Avenue F.D. Roosevelt 50 CP165/56, 1050, Brussels, Belgium
| | - Fuhong Su
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik, 808, 1070, Brussels, Belgium
| | - Jacques Creteur
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik, 808, 1070, Brussels, Belgium
| | - Fabio Silvio Taccone
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik, 808, 1070, Brussels, Belgium
| | - Nicolas Gaspard
- Department of Neurology, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik, 808, 1070, Brussels, Belgium.
| |
Collapse
|
13
|
Ghorbanzadeh V, Jafarpour A, Pirnia A, Pajouhi N, Khaksarian M, Veiskarami S, Nazari A. The role of vasopressin V1A and oxytocin OTR receptors in protective effects of arginine vasopressin against H 2O 2-induced oxidative stress in H9C2 cells. Arch Physiol Biochem 2022; 128:830-835. [PMID: 32141340 DOI: 10.1080/13813455.2020.1729816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Oxidative stress, has been shown to play an important role in the pathophysiology of cardiac remodelling and heart failure. The aim of study is effect of arginine vasopressin (AVP) on apoptosis of cardiomyocyte via its receptors. MATERIALS AND METHODS The cell viability effect of AVP in H9C2 cardiomyocytes was assayed using the MTT method. The transcription and translation level of apoptosis genes (Bax, Bcl-2, caspase-3) were discovered with qRT-PCR and western blotting. RESULTS The results showed that vasopressin could reduce apoptosis in cardiomyocytes cell line through downregulation of caspase-3, BAX and upregulation of Bcl-2 (p < .001). Also, there was a decrease in anti-apoptosis effect of vasopressin when V1A and OTR receptors were blocked with their antagonists. DISCUSSION These results suggest that activation of V1A and OTR receptors in H9C2 cells mediate protective effect of vasopressin via regulating apoptosis marker that lead to cell survival under conditions of stress oxidative.Key pointAVP may contribute to the improvement of heart ischaemia through its actions on V1A and OTR receptors.
Collapse
Affiliation(s)
- Vajihe Ghorbanzadeh
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences Khorramabad, Iran
| | - Afsaneh Jafarpour
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Afshin Pirnia
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences Khorramabad, Iran
| | - Naser Pajouhi
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences Khorramabad, Iran
- Department of Physiology, Lorestan University of Medical Science, Khorramabad, Iran
| | - Mojtaba Khaksarian
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences Khorramabad, Iran
- Department of Physiology, Lorestan University of Medical Science, Khorramabad, Iran
| | - Saeed Veiskarami
- Department of animal science, Lorestan Agricultural and Natural Resources Research and Education Center, Khorramabad, Iran
| | - Afshin Nazari
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences Khorramabad, Iran
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
- Department of Physiology, Lorestan University of Medical Science, Khorramabad, Iran
| |
Collapse
|
14
|
Rajanathan R, Pedersen TM, Thomsen MB, Botker HE, Matchkov VV. Phenylephrine-Induced Cardiovascular Changes in the Anesthetized Mouse: An Integrated Assessment of in vivo Hemodynamics Under Conditions of Controlled Heart Rate. Front Physiol 2022; 13:831724. [PMID: 35250634 PMCID: PMC8891648 DOI: 10.3389/fphys.2022.831724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Investigating the cardiovascular system is challenging due to its complex regulation by humoral and neuronal factors. Despite this complexity, many existing research methods are limited to the assessment of a few parameters leading to an incomplete characterization of cardiovascular function. Thus, we aim to establish a murine in vivo model for integrated assessment of the cardiovascular system under conditions of controlled heart rate. Utilizing this model, we assessed blood pressure, cardiac output, stroke volume, total peripheral resistance, and electrocardiogram (ECG). Hypothesis We hypothesize that (i) our in vivo model can be utilized to investigate cardiac and vascular responses to pharmacological intervention with the α1-agonist phenylephrine, and (ii) we can study cardiovascular function during artificial pacing of the heart, modulating cardiac function without a direct vascular effect. Methods We included 12 mice that were randomly assigned to either vehicle or phenylephrine intervention through intraperitoneal administration. Mice were anesthetized with isoflurane and intubated endotracheally for mechanical ventilation. We measured blood pressure via a solid-state catheter in the aortic arch, blood flow via a probe on the ascending aorta, and ECG from needle electrodes on the extremities. Right atrium was electrically paced at a frequency ranging from 10 to 11.3 Hz before and after either vehicle or phenylephrine administration. Results Phenylephrine significantly increased blood pressure, stroke volume, and total peripheral resistance compared to the vehicle group. Moreover, heart rate was significantly decreased following phenylephrine administration. Pacing significantly decreased stroke volume and cardiac output both prior to and after drug administration. However, phenylephrine-induced changes in blood pressure and total peripheral resistance were maintained with increasing pacing frequencies compared to the vehicle group. Total peripheral resistance was not significantly altered with increasing pacing frequencies suggesting that the effect of phenylephrine is primarily of vascular origin. Conclusion In conclusion, this in vivo murine model is capable of distinguishing between changes in peripheral vascular and cardiac functions. This study underlines the primary effect of phenylephrine on vascular function with secondary changes to cardiac function. Hence, this in vivo model is useful for the integrated assessment of the cardiovascular system.
Collapse
Affiliation(s)
- Rajkumar Rajanathan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- *Correspondence: Rajkumar Rajanathan,
| | | | - Morten B. Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Erik Botker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
15
|
Saghafi F, Bagheri N, Salehi-Abargouei A, Sahebnasagh A. Efficacy of combination triple therapy with vasopressin, steroid, and epinephrine in cardiac arrest: a systematic review and meta-analysis of randomized-controlled trials. J Intensive Care 2022; 10:5. [PMID: 35109925 PMCID: PMC8809021 DOI: 10.1186/s40560-022-00597-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 01/16/2022] [Indexed: 11/10/2022] Open
Abstract
Background This study investigated whether combination therapy with vasopressin, steroid, and epinephrine (VSE) improves in-hospital survival and return of spontaneous circulation (ROSC) during and after resuscitation in-hospital cardiac arrest (CA). Materials and methods Various databases were explored from inception until October 2021 for relevant published clinical trials and cohort studies. Results Three clinical trials were included. Pooled analysis suggested that VSE was significantly associated with increased ROSC in patients with in-hospital CA (IHCA) (odds ratio (OR): 2.281, 95% confidence interval (CI): 1.304–3.989, P value = 0.004). Meta-analysis of two studies (368 patients) demonstrated a significant difference in the reduction of mean arterial pressure (MAP) during and 15–20 min after cardiopulmonary resuscitation (standardized mean difference (SMD): 1.069, 95% CI: 0.851–1.288, P value < 0.001), renal failure free days (SMD = 0.590; 95% CI: 0.312–0.869 days; P value < 0.001), and coagulation failure free days (SMD = 0.403; 95% CI: 0.128–0.679, P value = 0.004). However, no significant difference was observed for survival-to-discharge ratio (OR: 2.082, 95% CI: 0.638–6.796, P value = 0.225) and ventilator free days (SMD = 0.201, 95% CI: − 0.677, 1.079 days; P value = 0.838). Conclusions VSE combination therapy during and after IHCA may have beneficial effects in terms of the ROSC, renal and circulatory failure free days, and MAP. Prospero registration: CRD42020178297 (05/07/2020). Supplementary Information The online version contains supplementary material available at 10.1186/s40560-022-00597-5.
Collapse
Affiliation(s)
- Fatemeh Saghafi
- Department of Clinical Pharmacy, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Negar Bagheri
- Pharmaceutical Sciences Research Center, School of Pharmacy, Student Research Committee, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Amin Salehi-Abargouei
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Adeleh Sahebnasagh
- Clinical Research Center, Department of Internal Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| |
Collapse
|
16
|
Myocardial Afterload Is a Key Biomechanical Regulator of Atrioventricular Myocyte Differentiation in Zebrafish. J Cardiovasc Dev Dis 2022; 9:jcdd9010022. [PMID: 35050232 PMCID: PMC8779957 DOI: 10.3390/jcdd9010022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 02/05/2023] Open
Abstract
Heart valve development is governed by both genetic and biomechanical inputs. Prior work has demonstrated that oscillating shear stress associated with blood flow is required for normal atrioventricular (AV) valve development. Cardiac afterload is defined as the pressure the ventricle must overcome in order to pump blood throughout the circulatory system. In human patients, conditions of high afterload can cause valve pathology. Whether high afterload adversely affects embryonic valve development remains poorly understood. Here we describe a zebrafish model exhibiting increased myocardial afterload, caused by vasopressin, a vasoconstrictive drug. We show that the application of vasopressin reliably produces an increase in afterload without directly acting on cardiac tissue in zebrafish embryos. We have found that increased afterload alters the rate of growth of the cardiac chambers and causes remodeling of cardiomyocytes. Consistent with pathology seen in patients with clinically high afterload, we see defects in both the form and the function of the valve leaflets. Our results suggest that valve defects are due to changes in atrioventricular myocyte signaling, rather than pressure directly acting on the endothelial valve leaflet cells. Cardiac afterload should therefore be considered a biomechanical factor that particularly impacts embryonic valve development.
Collapse
|
17
|
Abstract
Vasopressor use in severely injured trauma patients is discouraged due to concerns that vasoconstriction will worsen organ perfusion and result in increased mortality and organ failure in hypotensive trauma patients. Hypotensive resuscitation is advocated based on limited data that lower systolic blood pressure and mean arterial pressure will result in improved mortality. It is classically taught that hypotension and hypovolemia in trauma are associated with peripheral vasoconstriction. However, the pathophysiology of traumatic shock is complex and involves multiple neurohormonal interactions that are ultimately manifested by an initial sympathoexcitatory phase that attempts to compensate for acute blood loss and is characterized by vasoconstriction, tachycardia, and preserved mean arterial blood pressure. The subsequent hypotension observed in hemorrhagic shock reflects a sympathoinhibitory vasodilation phase. The objectives of hemodynamic resuscitation in hypotensive trauma patients are restoring adequate intravascular volume with a balanced ratio of blood products, correcting pathologic coagulopathy, and maintaining organ perfusion. Persistent hypotension and hypoperfusion are associated with worse coagulopathy and organ function. The practice of hypotensive resuscitation would appear counterintuitive to the goals of traumatic shock resuscitation and is not supported by consistent clinical data. In addition, excessive volume resuscitation is associated with adverse clinical outcomes. Therefore, in the resuscitation of traumatic shock, it is necessary to target an appropriate balance with intravascular volume and vascular tone. It would appear logical that vasopressors may be useful in traumatic shock resuscitation to counteract vasodilation in hemorrhage as well as other clinical conditions such as traumatic brain injury, spinal cord injury, multiple organ dysfunction syndrome, and vasodilation of general anesthetics. The purpose of this article is to discuss the controversy of vasopressors in hypotensive trauma patients and advocate for a nuanced approach to vasopressor administration in the resuscitation of traumatic shock.
Collapse
|
18
|
Avci II, Sahin I, Gungor B, Kücük SH, Sigirci S, Varol S, Tugrul S, Atici A, İnce O, Okuyan E. Higher Copeptin Levels are Associated with the Risk of Atrial Fibrillation in Patients with Rheumatic Mitral Stenosis. ACTA CARDIOLOGICA SINICA 2021; 37:412-419. [PMID: 34257491 PMCID: PMC8261697 DOI: 10.6515/acs.202107_37(4).20210111a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 01/11/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Correlations between increased copeptin levels and various cardiovascular diseases have been described. In this study, we aimed to investigate the correlation between increased copeptin levels and paroxysmal atrial fibrillation (PAF) in rheumatic mitral stenosis (MS). METHODS Patients with mild/moderate rheumatic MS and sinus rhythm were consecutively recruited from an echocardiography laboratory. Patients with a history of PAF and those with PAF on 24-48-hour ambulatory electrocardiography (ECG) monitoring constituted the study group, and those without PAF on ambulatory ECG monitoring constituted the control group. Clinical characteristics, echocardiographic parameters and levels of copeptin, plasma N-terminal proBNP (NT-proBNP) and high-sensitivity C-reactive protein (hs-CRP) were evaluated. RESULTS Twenty-nine patients with PAF and 124 control MS patients were studied. Patients in the PAF group were older, but the mitral valve areas and transmitral gradients were not different between the groups. In the PAF group, hs-CRP (1.2 vs. 0.8 mg/L, p < 0.001), NT-proBNP (335 vs. 115 pg/mL, p < 0.001) and copeptin (6.9 vs. 4.0 pmol/L, p < 0.001) levels were significantly higher than in the control group. Multivariable logistic regression analysis revealed that age [odds ratio (OR) 1.19, 95% confidence interval (CI) 1.04-1.38; p = 0.024], left atrial volume index (OR 1.23, 95% CI 1.06-1.41; p = 0.032), copeptin levels (OR 2.81, 95% CI 1.30-5.29; p < 0.001) and hs-CRP levels (OR 15.5, 95% CI 1.41-71.5; p = 0.012) were independent predictors of PAF. CONCLUSIONS In patients with mild/moderate rheumatic MS, higher copeptin and hs-CRP levels predicted a higher risk of developing atrial fibrillation.
Collapse
Affiliation(s)
- Ilhan Ilker Avci
- Department of Cardiology, Dr Siyami Ersek Thoracic and Cardiovascular Surgery Training and Research Hospital
| | | | - Baris Gungor
- Department of Cardiology, Dr Siyami Ersek Thoracic and Cardiovascular Surgery Training and Research Hospital
| | - Suat Hayri Kücük
- Department of Biochemistry, Bagcilar Training and Research Hospital
| | - Serhat Sigirci
- Department of Cardiology, Şişli Hamidiye Etfal Training and Research Hospital
| | | | | | - Adem Atici
- Department of Cardiology, Medeniyet Üniversitesi Göztepe Etfal Training and Research Hospital, Istanbul, Turkey
| | | | | |
Collapse
|
19
|
O'Connor KM, Ashoori M, Dias ML, Dempsey EM, O'Halloran KD, McDonald FB. Influence of innate immune activation on endocrine and metabolic pathways in infancy. Am J Physiol Endocrinol Metab 2021; 321:E24-E46. [PMID: 33900849 DOI: 10.1152/ajpendo.00542.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prematurity is the leading cause of neonatal morbidity and mortality worldwide. Premature infants often require extended hospital stays, with increased risk of developing infection compared with term infants. A picture is emerging of wide-ranging deleterious consequences resulting from innate immune system activation in the newborn infant. Those who survive infection have been exposed to a stimulus that can impose long-lasting alterations into later life. In this review, we discuss sepsis-driven alterations in integrated neuroendocrine and metabolic pathways and highlight current knowledge gaps in respect of neonatal sepsis. We review established biomarkers for sepsis and extend the discussion to examine emerging findings from human and animal models of neonatal sepsis that propose novel biomarkers for early identification of sepsis. Future research in this area is required to establish a greater understanding of the distinct neonatal signature of early and late-stage infection, to improve diagnosis, curtail inappropriate antibiotic use, and promote precision medicine through a biomarker-guided empirical and adjunctive treatment approach for neonatal sepsis. There is an unmet clinical need to decrease sepsis-induced morbidity in neonates, to limit and prevent adverse consequences in later life and decrease mortality.
Collapse
Affiliation(s)
- K M O'Connor
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - M Ashoori
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
| | - M L Dias
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - E M Dempsey
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, School of Medicine, College of Medicine and Health, Cork University Hospital, Wilton, Cork, Ireland
| | - K D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
| | - F B McDonald
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
| |
Collapse
|
20
|
Okuma Y, Aoki T, Miyara SJ, Hayashida K, Nishikimi M, Takegawa R, Yin T, Kim J, Becker LB, Shinozaki K. The evaluation of pituitary damage associated with cardiac arrest: An experimental rodent model. Sci Rep 2021; 11:629. [PMID: 33436714 PMCID: PMC7804952 DOI: 10.1038/s41598-020-79780-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 11/24/2020] [Indexed: 11/11/2022] Open
Abstract
The pituitary gland plays an important endocrinal role, however its damage after cardiac arrest (CA) has not been well elucidated. The aim of this study was to determine a pituitary gland damage induced by CA. Rats were subjected to 10-min asphyxia and cardiopulmonary resuscitation (CPR). Immunohistochemistry and ELISA assays were used to evaluate the pituitary damage and endocrine function. Samples were collected at pre-CA, and 30 and 120 min after cardio pulmonary resuscitation. Triphenyltetrazolium chloride (TTC) staining demonstrated the expansion of the pituitary damage over time. There was phenotypic validity between the pars distalis and nervosa. Both CT-proAVP (pars nervosa hormone) and GH/IGF-1 (pars distalis hormone) decreased over time, and a different expression pattern corresponding to the damaged areas was noted (CT-proAVP, 30.2 ± 6.2, 31.5 ± 5.9, and 16.3 ± 7.6 pg/mg protein, p < 0.01; GH/IGF-1, 2.63 ± 0.61, 0.62 ± 0.36, and 2.01 ± 0.41 ng/mg protein, p < 0.01 respectively). Similarly, the expression pattern between these hormones in the end-organ systems showed phenotypic validity. Plasma CT-proAVP (r = 0.771, p = 0.025) and IGF-1 (r = −0.775, p = 0.024) demonstrated a strong correlation with TTC staining area. Our data suggested that CA induces pathological and functional damage to the pituitary gland.
Collapse
Affiliation(s)
- Yu Okuma
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Tomoaki Aoki
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Santiago J Miyara
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, USA
| | - Kei Hayashida
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Mitsuaki Nishikimi
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Ryosuke Takegawa
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Tai Yin
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Junhwan Kim
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA
| | - Lance B Becker
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA.,Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Koichiro Shinozaki
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY, 11030, USA. .,Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
21
|
da Costa LHA, Santos-Junior NN, Catalão CHR, Rocha MJA. Microglial Activation Modulates Neuroendocrine Secretion During Experimental Sepsis. Mol Neurobiol 2021; 58:2133-2144. [PMID: 33415683 DOI: 10.1007/s12035-020-02241-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/26/2020] [Indexed: 01/02/2023]
Abstract
Sepsis promotes an inflammatory state in the central nervous system (CNS) that may cause autonomic, cognitive, and endocrine changes. Microglia, a resident immune cell of the CNS, is activated in several brain regions during sepsis, suggesting its participation in the central alterations observed in this disease. In this study, we aimed to investigate the role of microglial activation in the neuroendocrine system functions during systemic inflammation. Wistar rats received an intracerebroventricular injection of the microglial activation inhibitor minocycline (100 μg/animal), shortly before sepsis induction by cecal ligation and puncture. At 6 and 24 h after surgery, hormonal parameters, central and peripheral inflammation, and markers of apoptosis and synaptic function in the hypothalamus were analyzed. The administration of minocycline decreased the production of inflammatory mediators and the expression of cell death markers, especially in the late phase of sepsis (24 h). With respect to the endocrine parameters, microglial inhibition caused a decrease in oxytocin and an increase in corticosterone and vasopressin plasma levels in the early phase of sepsis (6 h), while in the late phase, we observed decreased oxytocin and increased ACTH and corticosterone levels compared to septic animals that did not receive minocycline. Prolactin levels were not affected by minocycline administration. The results indicate that microglial activation differentially modulates the secretion of several hormones and that this process is associated with inflammatory mediators produced both centrally and peripherally.
Collapse
Affiliation(s)
- Luis Henrique Angenendt da Costa
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Nilton Nascimento Santos-Junior
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Carlos Henrique Rocha Catalão
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Maria José Alves Rocha
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil. .,Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14040-904, Brazil. .,Departamento de Biologia Básica e Oral, Faculdade de Odontologia de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n, Ribeirão Preto, SP, 14040-904, Brazil.
| |
Collapse
|
22
|
Alsaadi AS, Sushko K, Bui V, Van Den Anker J, Razak A, Samiee-Zafarghandy S. Efficacy and safety of vasopressin and terlipressin in preterm neonates: a protocol for a systematic review. BMJ Paediatr Open 2021; 5:e001067. [PMID: 34179513 PMCID: PMC8191613 DOI: 10.1136/bmjpo-2021-001067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/13/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The use of vasoactive agents like arginine vasopressin (AVP) and terlipressin to treat hypotension or persistent pulmonary hypertension in critically ill preterm neonates is increasing. Therefore, a systematic review of the available data on dosing, efficacy and safety of AVP and terlipressin in this patient population appears beneficial. METHODS We will conduct a systematic review of the available evidence on the use of AVP and terlipressin for the treatment of hypotension or persistent pulmonary hypertension in preterm neonates. We will search Ovid MEDLINE, EMBASE, the Cochrane Central Register of Controlled Trials, Web of Science and Google Scholar from inception to March 2021. Two reviewers will independently screen titles and abstracts, review the full text of eligible studies, extract data, assess the risk of bias and judge the certainty of the evidence. Our primary outcome will be an (1) improvement of end-organ perfusion after initiation of AVP or terlipressin and (2) mortality prior to discharge. Our secondary outcomes will include (1) major neurosensory abnormality and (2) the occurrence of adverse events. DISCUSSION The currently available evidence on the efficacy and safety of AVP and terlipressin in preterm neonates is limited. Yet, evidence on the pharmacology of these drugs and the pathophysiology of vasoplegic shock support the biological plausibility for their clinical effectiveness in this population. Therefore, we aim to address this gap concerning the use of vasopressin and terlipressin among critically ill preterm neonates. TRIAL REGISTRATION This protocol has been submitted for registration to the international database of prospectively registered systematic reviews (PROSPERO, awaiting registration number).
Collapse
Affiliation(s)
| | - Katelyn Sushko
- Faculty of Health Sciences, School of Nursing, McMaster University, Hamilton, Ontario, Canada
| | - Vivian Bui
- Department of Pharmacy, Hamilton, Ontario, Canada
| | - John Van Den Anker
- Department of Pediatrics and Pharmacology and Pharmacometric, University Children's Hospital Basel, Basel, Switzerland.,Division of Clinical Pharmacology, Children's National Hospital, Washintgon, DC, USA.,Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital, Rotterdam, Netherlands
| | - Abdul Razak
- Department of Pediatrics, Princess Norah Bint Abdulrahman University, Riyadh, Saudi Arabia.,Division of Neonatology, Department of Pediatrics, King Abdullah bin Abdulaziz University Hospital, King Abdullah bin Abdulaziz University Hospital, Riyadh, Saudi Arabia
| | | |
Collapse
|
23
|
Boyd SM, Riley KL, Giesinger RE, McNamara PJ. Use of vasopressin in neonatal hypertrophic obstructive cardiomyopathy: case series. J Perinatol 2021; 41:126-133. [PMID: 32951013 DOI: 10.1038/s41372-020-00824-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/17/2020] [Accepted: 09/11/2020] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To determine the effect of vasopressin on arterial blood pressure in infants with neonatal hypertrophic obstructive cardiomyopathy (HOCM). STUDY DESIGN Retrospective case study in Neonatal ICU involving six infants; five born to mothers with diabetes mellitus (mean gestational age 37.5 ± 0.9 weeks). Vasopressin infusion was started at a mean dose of 0.3 ± 0.2 mU/kg/min. RESULT Initiation of vasopressin was followed by improved mean (p = 0.004), systolic (p = 0.028), and diastolic (p = 0.009) arterial pressure within 2 h. Heart rate (p = 0.025) and oxygen requirement (p = 0.021) also declined after initiation. Serum sodium declined initially and recovered by 72 h (p = 0.017). CONCLUSION Although there is limited experience with vasopressin use in neonatal HOCM, our case series suggests it may be beneficial for improving systemic hypotension and stabilization of hemodynamics. The potential for hyponatremia is high, necessitating careful fluid/electrolyte management. A prospective randomized trial is necessary to confirm safety and efficacy of vasopressin treatment in neonatal HOCM.
Collapse
Affiliation(s)
- Stephanie M Boyd
- Neonatology Department, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G1X8, Canada.,Grace Centre for Newborn Intensive Care, Children's Hospital at Westmead, Cnr Hawkesbury Rd and Hainsworth St, Westmead, NSW, 2145, Australia
| | - Kristin L Riley
- Division of Neonatology, The University of Iowa, 200 Hawkins Dr, Iowa City, IA, 52242, USA
| | - Regan E Giesinger
- Neonatology Department, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G1X8, Canada.,Division of Neonatology, The University of Iowa, 200 Hawkins Dr, Iowa City, IA, 52242, USA
| | - Patrick J McNamara
- Neonatology Department, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G1X8, Canada. .,Division of Neonatology, The University of Iowa, 200 Hawkins Dr, Iowa City, IA, 52242, USA.
| |
Collapse
|
24
|
Sims CA, Holena D, Kim P, Pascual J, Smith B, Martin N, Seamon M, Shiroff A, Raza S, Kaplan L, Grill E, Zimmerman N, Mason C, Abella B, Reilly P. Effect of Low-Dose Supplementation of Arginine Vasopressin on Need for Blood Product Transfusions in Patients With Trauma and Hemorrhagic Shock: A Randomized Clinical Trial. JAMA Surg 2020; 154:994-1003. [PMID: 31461138 DOI: 10.1001/jamasurg.2019.2884] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Current therapies for traumatic blood loss focus on hemorrhage control and blood volume replacement. Severe hemorrhagic shock, however, is associated with a state of arginine vasopressin (AVP) deficiency, and supplementation of this hormone may decrease the need for blood products in resuscitation. Objective To determine whether low-dose supplementation of AVP in patients with trauma (hereinafter referred to as trauma patients) and with hemorrhagic shock decreases their need for transfused blood products during resuscitation. Design, Setting, and Participants This randomized, double-blind placebo-controlled clinical trial included adult trauma patients (aged 18-65 years) who received at least 6 U of any blood product within 12 hours of injury at a single urban level 1 trauma center from May 1, 2013, through May 31, 2017. Exclusion criteria consisted of prehospital cardiopulmonary resuscitation, emergency department thoracotomy, corticosteroid use, chronic renal insufficiency, coronary artery disease, traumatic brain injury requiring any neurosurgical intervention, pregnancy, prisoner status, or AVP administration before enrollment. Data were analyzed from May 1, 2013, through May 31, 2017, using intention to treat and per protocol. Interventions After administration of an AVP bolus (4 U) or placebo, participants received AVP (≤0.04 U/min) or placebo for 48 hours to maintain a mean arterial blood pressure of at least 65 mm Hg. Main Outcomes The primary outcome was total volume of blood product transfused. Secondary end points included total volume of crystalloid transfused, vasopressor requirements, secondary complications, and 30-day mortality. Results One hundred patients underwent randomization (49 to the AVP group and 51 to the placebo group). Patients were primarily young (median age, 27 years [interquartile range {IQR}, 22-25 years]) and male (n = 93) with penetrating trauma (n = 79). Cohort characteristics before randomization were well balanced. At 48 hours, patients who received AVP required significantly less blood products (median, 1.4 [IQR, 0.5-2.6] vs 2.9 [IQR, 1.1-4.8] L; P = .01) but did not differ in requirements for crystalloids (median, 9.9 [IQR, 7.9-13.0] vs 11.0 [8.9-15.0] L; P = .22) or vasopressors (median, 400 [IQR, 0-5900] vs 1400 [IQR, 200-7600] equivalent units; P = .22). Although the groups had similar rates of mortality (6 of 49 [12%] vs 6 of 51 [12%]; P = .94) and total complications (24 of 44 [55%] vs 30 of 47 [64%]; P = .37), the AVP group had less deep venous thrombosis (5 of 44 [11%] vs 16 of 47 [34%]; P = .02). Conclusions and Relevance Low-dose AVP during the resuscitation of trauma patients in hemorrhagic shock decreases blood product requirements. Additional research is necessary to determine whether including AVP improves morbidity or mortality. Trial Registration ClinicalTrials.gov identifier: NCT01611935.
Collapse
Affiliation(s)
- Carrie A Sims
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia.,Penn Acute Research Collaboration (PARC), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Daniel Holena
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Patrick Kim
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia
| | - Jose Pascual
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia
| | - Brian Smith
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia
| | - Neils Martin
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia
| | - Mark Seamon
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia
| | - Adam Shiroff
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia
| | - Shariq Raza
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia
| | - Lewis Kaplan
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia
| | - Elena Grill
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia
| | - Nicole Zimmerman
- Department of Quantitative Health Sciences and Outcomes Research, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Christopher Mason
- Department of Anesthesia, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Benjamin Abella
- Department of Emergency Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Patrick Reilly
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Department of Surgery, University of Pennsylvania, Philadelphia
| |
Collapse
|
25
|
Masud F, Gheewala G, Giesecke M, Suarez EE, Ratnani I. Cardiogenic Shock in Perioperative and Intraoperative Settings: A Team Approach. Methodist Debakey Cardiovasc J 2020; 16:e1-e7. [PMID: 32280425 DOI: 10.14797/mdcj-16-1-e1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cardiogenic shock (CS) is a multifactorial disease process with high morbidity and mortality. When it occurs in a peri- or intraoperative setting, factors such as surgery, anesthesia, and post-surgical physiology can negatively affect patient outcomes. Since patient needs often escalate during CS-from medications to mechanical support to palliative care-this disease demands a multidisciplinary approach that encompasses all aspects of medical delivery. Preliminary studies have indicated that a multidisciplinary team approach to CS results in earlier diagnosis and treatment and improves patient outcomes. Here we discuss various management strategies for CS from an anesthesiology, surgery, and critical care perspective.
Collapse
Affiliation(s)
- Faisal Masud
- HOUSTON METHODIST DEBAKEY HEART & VASCULAR CENTER, HOUSTON METHODIST HOSPITAL, HOUSTON, TEXAS
| | - Gaurav Gheewala
- HOUSTON METHODIST DEBAKEY HEART & VASCULAR CENTER, HOUSTON METHODIST HOSPITAL, HOUSTON, TEXAS
| | - Martin Giesecke
- HOUSTON METHODIST DEBAKEY HEART & VASCULAR CENTER, HOUSTON METHODIST HOSPITAL, HOUSTON, TEXAS
| | - E E Suarez
- HOUSTON METHODIST DEBAKEY HEART & VASCULAR CENTER, HOUSTON METHODIST HOSPITAL, HOUSTON, TEXAS
| | - Iqbal Ratnani
- HOUSTON METHODIST DEBAKEY HEART & VASCULAR CENTER, HOUSTON METHODIST HOSPITAL, HOUSTON, TEXAS
| |
Collapse
|
26
|
Schultz J, Andersen A, Lyhne MD, Arcanjo DDR, Kjaergaard B, Simonsen U, Nielsen-Kudsk JE. Terlipressin Increases Systemic and Lowers Pulmonary Arterial Pressure in Experimental Acute Pulmonary Embolism. Crit Care Med 2020; 48:e308-e315. [PMID: 32205621 DOI: 10.1097/ccm.0000000000004243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES We investigated whether the vasopressin-analog, terlipressin induces systemic vasoconstriction and pulmonary vasodilation in a porcine model of acute pulmonary embolism. DESIGN Controlled, animal study. SETTING Tertiary medical center research laboratory. SUBJECTS Female pigs (n = 12, Cross of Land Race, Duroc, and Yorkshire ~ 60 kg). INTERVENTIONS Acute pulmonary embolism was induced by administration of three large autologous emboli. Animals then received four increasing doses of either terlipressin (n = 6) or vehicle (n = 6). MEASUREMENTS AND MAIN RESULTS Effects were evaluated in vivo at baseline, after pulmonary embolism and after each dose by invasive hemodynamic measures, transesophageal echocardiography, and blood analysis. Isolated pulmonary arteries were evaluated ex vivo in a myograph. Pulmonary embolism caused a four-fold increase in pulmonary vascular resistance (p < 0.0001) and a two-fold increase in mean pulmonary arterial pressure (p < 0.0001) compared with baseline. Terlipressin increased mean systemic blood pressure (28 ± 5 mm Hg; p < 0.0001) and systemic vascular resistance (1,320 ± 143 dynes; p < 0.0001) compared with vehicle. In the pulmonary circulation, terlipressin decreased mean pulmonary arterial pressure (-6.5 ± 1.8 mm Hg; p = 0.005) and tended to decrease pulmonary vascular resistance (-83 ± 33 dynes; p = 0.07). Terlipressin decreased cardiac output (-2.5 ± 0.5 L/min; p < 0.0001) and increased plasma lactate (2.7 ± 0.2 mmol/L; p < 0.0001), possibly indicating systemic hypoperfusion. A biomarker of cerebral ischemia, S100b, remained unchanged, suggesting preserved cerebral perfusion (0.17 ± 0.11 µg/L; p = 0.51). Ex vivo, terlipressin relaxed pulmonary and constricted mesenteric arteries. CONCLUSIONS Terlipressin caused systemic vasoconstriction and pulmonary vasodilation in a porcine in vivo model of acute pulmonary embolism and vasorelaxation in isolated pulmonary arteries. Despite positive vascular effects, cardiac output declined and plasma lactate increased probably due to a predominantly systemic vasoconstrictor effect of terlipressin. These findings should warrant careful translation to the clinical setting and does not suggest routine use in acute pulmonary embolism.
Collapse
Affiliation(s)
- Jacob Schultz
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Asger Andersen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Mads D Lyhne
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Daniel D R Arcanjo
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
- Institute of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Ulf Simonsen
- Institute of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jens Erik Nielsen-Kudsk
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
27
|
Carinci V, Gamberini L, Coniglio C, Casella G, Gordini G, Di Pasquale G. Catecholaminergic Polymorphic Ventricular Tachycardia: Challenges During Resuscitation and Post-Cardiac Arrest Care. J Emerg Med 2020; 58:677-681. [PMID: 32204998 DOI: 10.1016/j.jemermed.2020.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/28/2019] [Accepted: 01/12/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a rare channelopathy involving cardiac calcium metabolism that often shows up at an early age with misleading clinical symptoms, such as emotion- or exercise-related syncope with a normal resting electrocardiogram. In addition, it might be the underlying cause of sudden cardiac arrest in children or young adults. The particular pathophysiology of CPVT makes it particularly challenging for both resuscitation and the subsequent intensive care management after return of spontaneous circulation (ROSC). CASE REPORT We describe a case of sudden cardiac arrest in an 11-year-old girl affected by CPVT, with a particular focus on the most challenging aspects of resuscitation and intensive care management in light of other experiences found in the literature. A warning about the prodysrythmicity of mild hypothermia induced in the context of post-ROSC targeted temperature management in this particular population of patients and its possible physiopathological basis are discussed. WHY SHOULD AN EMERGENCY PHYSICIAN BE AWARE OF THIS?: CPVT is a rare but potentially lethal cause of stress-related syncope and sudden cardiac arrest in children and young adults. The diagnosis of CPVT requires a high level of suspicion and an interdisciplinary approach, including some adjustments during resuscitation and post-cardiac arrest care.
Collapse
Affiliation(s)
- Valeria Carinci
- Division of Cardiology, Ospedale Maggiore Carlo Alberto Pizzardi, Bologna, Italy
| | - Lorenzo Gamberini
- Division of Anesthesia, Intensive Care and Prehospital Emergency, Ospedale Maggiore Carlo Alberto Pizzardi, Bologna, Italy
| | - Carlo Coniglio
- Division of Anesthesia, Intensive Care and Prehospital Emergency, Ospedale Maggiore Carlo Alberto Pizzardi, Bologna, Italy
| | - Gianni Casella
- Division of Cardiology, Ospedale Maggiore Carlo Alberto Pizzardi, Bologna, Italy
| | - Giovanni Gordini
- Division of Anesthesia, Intensive Care and Prehospital Emergency, Ospedale Maggiore Carlo Alberto Pizzardi, Bologna, Italy
| | - Giuseppe Di Pasquale
- Division of Cardiology, Ospedale Maggiore Carlo Alberto Pizzardi, Bologna, Italy
| |
Collapse
|
28
|
|
29
|
Shimauchi T, Maki J, Yoshino J, Fujimura N, Hoka S. Effectiveness of arginine vasopressin for the management of refractory hemorrhagic shock in a patient with autonomic dysreflexia caused by spinal cord injury. JA Clin Rep 2018; 4:79. [PMID: 32026013 PMCID: PMC6966762 DOI: 10.1186/s40981-018-0216-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/01/2018] [Indexed: 11/18/2022] Open
Abstract
Background Arginine vasopressin has been used for the management of refractory vasodilatory shock. However, it is still unclear whether arginine vasopressin is useful for hypotension in patients with spinal cord injury. Case description A 78-year-old man with autonomic dysreflexia and paralysis below the level corresponding to Th2 due to spinal cord injury previously underwent cholecystectomy. During the surgery, accidental hemorrhage led him to refractory hemorrhagic shock unresponsive to fluid resuscitation and catecholamine. Lasting hypotension was improved with arginine vasopressin. Conclusion We described a rare case report on the use of arginine vasopressin for management of refractory hemorrhagic shock in a patient with autonomic dysreflexia.
Collapse
Affiliation(s)
- Tsukasa Shimauchi
- Operating Rooms, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Jun Maki
- Emergency and Critical Care Center, Kyushu University Hospital, Fukuoka, Japan
| | - Jun Yoshino
- Department of Anesthesiology, St Mary's Hospital, Kurume, Japan
| | | | - Sumio Hoka
- Department of Anesthesiology and Critical Care Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
30
|
Chirinos JA, Sardana M, Oldland G, Ansari B, Lee J, Hussain A, Mustafa A, Akers SR, Wei W, Lakatta EG, Fedorova OV. Association of arginine vasopressin with low atrial natriuretic peptide levels, left ventricular remodelling, and outcomes in adults with and without heart failure. ESC Heart Fail 2018; 5:911-919. [PMID: 29969536 PMCID: PMC6165935 DOI: 10.1002/ehf2.12319] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/30/2018] [Indexed: 01/14/2023] Open
Abstract
Aims The arginine vasopressin (AVP) pathway has been extensively studied in heart failure (HF) with reduced ejection fraction (HFrEF), but less is known about AVP in HF with preserved EF (HFpEF). Furthermore, the association between AVP and atrial natriuretic peptide (ANP, a well‐known inhibitor of AVP secretion) in HF is unknown. Methods and results We studied subjects with HFpEF (n = 28) and HFrEF (n = 25) and without HF (n = 71). Left ventricular (LV) mass and left atrial (LA) volumes were measured with cardiac magnetic resonance imaging. Arginine vasopressin and ANP were measured with enzyme‐linked immunosorbent assay. Arginine vasopressin levels were significantly greater in HFpEF [0.96 pg/mL; 95% confidence interval (CI) = 0.83–1.1 pg/mL] compared with subjects without HF (0.69 pg/mL; 95% CI = 0.6–0.77 pg/mL; P = 0.0002). Heart failure with preserved ejection fraction (but not HFrEF) was a significant predictor of higher AVP after adjustment for potential confounders. Arginine vasopressin levels were independently associated with a greater LA volume and also paradoxically, with lower ANP levels. Key independent correlates of higher AVP were the presence of HFpEF (standardized β = 0.32; 95% CI = 0.09–0.56; P = 0.0073) and the ANP/LA volume ratio (standardized β = −0.23; 95% CI = −0.42 to −0.04; P = 0.0196). Arginine vasopressin levels were independently associated with LV mass (β = 0.26; 95% CI = 0.09–0.43; P = 0.003) and with an increased risk of death or HF admissions during follow‐up (hazard ratio = 1.61; 95% CI = 1.13–2.29; P = 0.008). Conclusions Arginine vasopressin is increased in HFpEF and is associated with LV hypertrophy and poor outcomes. Higher AVP is associated with the combination of LA enlargement and paradoxically low ANP levels. These findings may indicate that a relative deficiency of ANP (an inhibitor of AVP secretion) in the setting of chronically increased LA pressure may contribute to AVP excess.
Collapse
Affiliation(s)
- Julio A Chirinos
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Mayank Sardana
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Garrett Oldland
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bilal Ansari
- Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan Lee
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Anila Hussain
- Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Anique Mustafa
- Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Scott R Akers
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, USA
| | - Wen Wei
- National Institute of Aging, Baltimore, MD, USA
| | | | | |
Collapse
|
31
|
Epigenetic Programming of Synthesis, Release, and/or Receptor Expression of Common Mediators Participating in the Risk/Resilience for Comorbid Stress-Related Disorders and Coronary Artery Disease. Int J Mol Sci 2018; 19:ijms19041224. [PMID: 29670001 PMCID: PMC5979500 DOI: 10.3390/ijms19041224] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/09/2018] [Accepted: 04/16/2018] [Indexed: 02/07/2023] Open
Abstract
Corticotrophin releasing factor, vasopressin, oxytocin, natriuretic hormones, angiotensin, neuregulins, some purinergic substances, and some cytokines contribute to the long-term modulation and restructuring of cardiovascular regulation networks and, at the same time, have relevance in situations of comorbid abnormal stress responses. The synthesis, release, and receptor expression of these mediators seem to be under epigenetic control since early stages of life, possibly underlying the comorbidity to coronary artery disease (CAD) and stress-related disorders (SRD). The exposure to environmental conditions, such as stress, during critical periods in early life may cause epigenetic programming modifying the development of pathways that lead to stable and long-lasting alterations in the functioning of these mediators during adulthood, determining the risk of or resilience to CAD and SRD. However, in contrast to genetic information, epigenetic marks may be dynamically altered throughout the lifespan. Therefore, epigenetics may be reprogrammed if the individual accepts the challenge to undertake changes in their lifestyle. Alternatively, epigenetics may remain fixed and/or even be inherited in the next generation. In this paper, we analyze some of the common neuroendocrine functions of these mediators in CAD and SRD and summarize the evidence indicating that they are under early programming to put forward the theoretical hypothesis that the comorbidity of these diseases might be epigenetically programmed and modified over the lifespan of the individual.
Collapse
|
32
|
Skrzypecki J, Grabska-Liberek I, Przybek J, Ufnal M. A common humoral background of intraocular and arterial blood pressure dysregulation. Curr Med Res Opin 2018; 34:521-529. [PMID: 29219620 DOI: 10.1080/03007995.2017.1415203] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND It has been postulated that intraocular pressure, an important glaucoma risk factor, correlates positively with arterial blood pressure (blood pressure). However, results of experimental and clinical studies are often contradictory. It is hypothesized that, in some hypertensive patients, disturbances in intraocular pressure regulation may depend on biological effects of blood borne hormones underlying a particular type of hypertension, rather than on blood pressure level itself. REVIEW This review compares the effects of hormones on blood pressure and intraocular pressure, in order to identify a hormonal profile of hypertensive patients with an increased risk of intraocular pressure surge. The PUBMED database was searched to identify pre-clinical and clinical studies investigating the role of angiotensin II, vasopressin, adrenaline, noradrenaline, prostaglandins, and gaseous transmitters in the regulation of blood pressure and intraocular pressure. RESULTS Studies included in the review suggest that intraocular and blood pressures often follow a different pattern of response to the same hormone. For example, vasopressin increases blood pressure, but decreases intraocular pressure. In contrast, high level of nitric oxide decreases blood pressure, but increases intraocular pressure. CONCLUSIONS Arterial hypertension is associated with altered levels of blood borne hormones. Contradicting results of studies on the relationship between arterial hypertension and intraocular pressure might be partially explained by diverse effects of hormones on arterial and intraocular pressures. Further studies are needed to evaluate if hormonal profiling may help to identify glaucoma-prone patients.
Collapse
Affiliation(s)
- Janusz Skrzypecki
- a Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research , Medical University of Warsaw , Warsaw , Poland
- b Department of Ophthalmology , Medical Center for Postgraduate Education , Warsaw , Poland
| | - Iwona Grabska-Liberek
- b Department of Ophthalmology , Medical Center for Postgraduate Education , Warsaw , Poland
| | - Joanna Przybek
- c Department of Experimental and Clinical Pharmacology , Medical University of Warsaw , Poland
| | - Marcin Ufnal
- a Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research , Medical University of Warsaw , Warsaw , Poland
| |
Collapse
|
33
|
Muttenthaler M, Andersson Å, Vetter I, Menon R, Busnelli M, Ragnarsson L, Bergmayr C, Arrowsmith S, Deuis JR, Chiu HS, Palpant NJ, O'Brien M, Smith TJ, Wray S, Neumann ID, Gruber CW, Lewis RJ, Alewood PF. Subtle modifications to oxytocin produce ligands that retain potency and improved selectivity across species. Sci Signal 2017; 10:10/508/eaan3398. [PMID: 29208680 DOI: 10.1126/scisignal.aan3398] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oxytocin and vasopressin mediate various physiological functions that are important for osmoregulation, reproduction, cardiovascular function, social behavior, memory, and learning through four G protein-coupled receptors that are also implicated in high-profile disorders. Targeting these receptors is challenging because of the difficulty in obtaining ligands that retain selectivity across rodents and humans for translational studies. We identified a selective and more stable oxytocin receptor (OTR) agonist by subtly modifying the pharmacophore framework of human oxytocin and vasopressin. [Se-Se]-oxytocin-OH displayed similar potency to oxytocin but improved selectivity for OTR, an effect that was retained in mice. Centrally infused [Se-Se]-oxytocin-OH potently reversed social fear in mice, confirming that this action was mediated by OTR and not by V1a or V1b vasopressin receptors. In addition, [Se-Se]-oxytocin-OH produced a more regular contraction pattern than did oxytocin in a preclinical labor induction and augmentation model using myometrial strips from cesarean sections. [Se-Se]-oxytocin-OH had no activity in human cardiomyocytes, indicating a potentially improved safety profile and therapeutic window compared to those of clinically used oxytocin. In conclusion, [Se-Se]-oxytocin-OH is a novel probe for validating OTR as a therapeutic target in various biological systems and is a promising new lead for therapeutic development. Our medicinal chemistry approach may also be applicable to other peptidergic signaling systems with similar selectivity issues.
Collapse
Affiliation(s)
- Markus Muttenthaler
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia. .,Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Åsa Andersson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.,School of Pharmacy, The University of Queensland, Brisbane, Queensland 4104, Australia
| | - Rohit Menon
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, 93053 Regensburg, Germany
| | - Marta Busnelli
- CNR-Institute of Neuroscience, 20129 Milan, Italy.,Department of Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Lotten Ragnarsson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Christian Bergmayr
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Sarah Arrowsmith
- Department of Cellular and Molecular Physiology, Harris-Wellbeing Preterm Birth Centre, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK
| | - Jennifer R Deuis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Han Sheng Chiu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Nathan J Palpant
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Margaret O'Brien
- National Centre for Biomedical Engineering Science, National University of Ireland, Galway H91 CF50, Ireland
| | - Terry J Smith
- National Centre for Biomedical Engineering Science, National University of Ireland, Galway H91 CF50, Ireland
| | - Susan Wray
- Department of Cellular and Molecular Physiology, Harris-Wellbeing Preterm Birth Centre, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK
| | - Inga D Neumann
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, 93053 Regensburg, Germany
| | - Christian W Gruber
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria.,School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Paul F Alewood
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
34
|
Sims CA, Yuxia G, Singh K, Werlin EC, Reilly PM, Baur JA. Supplemental arginine vasopressin during the resuscitation of severe hemorrhagic shock preserves renal mitochondrial function. PLoS One 2017; 12:e0186339. [PMID: 29065123 PMCID: PMC5655425 DOI: 10.1371/journal.pone.0186339] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 10/01/2017] [Indexed: 01/30/2023] Open
Abstract
Arginine vasopressin (AVP), a hormone secreted by the posterior pituitary, plays a vital role in maintaining vasomotor tone during acute blood loss. We hypothesized that decompensated hemorrhagic shock is associated with decreased AVP stores and supplementation during resuscitation would improve both blood pressure and renal function. Using a decompensated hemorrhagic shock model, male Long-Evans rats were bled to mean arterial blood pressure (MAP) of 40mmHg and maintained until the MAP could not be sustained without fluid. Once 40% of the shed volume was returned in lactated Ringer’s (Severe Shock), animals were resuscitated over 60 minutes with 4x the shed volume in lactated Ringer’s (LR) or the same fluids with AVP (0.5 units/kg+ 0.03 units/kg/min). Animals (n = 6-9/group) were sacrificed before hemorrhage (Sham), at Severe Shock, following resuscitation (60R, 60R with AVP) or 18 hours post-resuscitation (18hr, 18hr with AVP). Blood samples were taken to measure AVP levels and renal function. Pituitaries were harvested and assayed for AVP. Kidney samples were taken to assess mitochondrial function, histology, and oxidative damage. Baseline pituitary AVP stores (30,364 ± 5311 pg/mg) decreased with severe shock and were significantly depressed post-resuscitation (13,910 ± 3016 pg/ml. p<0.05) and at 18hr (15,592 ±1169 pg/ml, p<0.05). Resuscitation with LR+AVP led to higher serum AVP levels at 60R (31±8 vs 79±12; p<0.01) with an improved MAP both at 60R (125±3 vs 77±7mmHg; p<0.01) and 18hr (82±6 vs 69±5mmHg;p<0.05). AVP supplementation preserved complex I respiratory capacity at 60R and both complex I and II function at 18hr (p<0.05). AVP was also associated with decreased reactive oxygen species at 60R (856±67 vs 622±48F RFU) and significantly decreased oxidative damage as measured by mitochondrial lipid peroxidation (0.9±0.1 vs 1.7±0.1 fold change, p<0.01) and nitrosylation (0.9±0.1 vs 1.4±0.2 fold change, p<0.05). With AVP, renal damage was mitigated at 60R and histologic architecture was conserved at 18 hours. In conclusion, pituitary and serum AVP levels decrease during severe hemorrhage and may contribute to the development of decompensated hemorrhagic shock. Supplementing exogenous AVP during resuscitation improves blood pressure, preserves renal mitochondrial function, and mitigates acute kidney injury.
Collapse
Affiliation(s)
- Carrie A. Sims
- The Trauma Center at the University of Pennsylvania, Department of Surgery, Perelman School of Medicine, Philadelphia, PA, United States of America
- Penn Acute Research Collaboration (PARC), University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail:
| | - Guan Yuxia
- The Trauma Center at the University of Pennsylvania, Department of Surgery, Perelman School of Medicine, Philadelphia, PA, United States of America
| | - Khushboo Singh
- The Trauma Center at the University of Pennsylvania, Department of Surgery, Perelman School of Medicine, Philadelphia, PA, United States of America
| | - Evan C. Werlin
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States of America
| | - Patrick M. Reilly
- The Trauma Center at the University of Pennsylvania, Department of Surgery, Perelman School of Medicine, Philadelphia, PA, United States of America
| | - Joseph A. Baur
- Penn Acute Research Collaboration (PARC), University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| |
Collapse
|
35
|
Burford NG, Webster NA, Cruz-Topete D. Hypothalamic-Pituitary-Adrenal Axis Modulation of Glucocorticoids in the Cardiovascular System. Int J Mol Sci 2017; 18:ijms18102150. [PMID: 29035323 PMCID: PMC5666832 DOI: 10.3390/ijms18102150] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/06/2017] [Accepted: 10/11/2017] [Indexed: 02/06/2023] Open
Abstract
The collective of endocrine organs acting in homeostatic regulation—known as the hypothalamic-pituitary-adrenal (HPA) axis—comprises an integration of the central nervous system as well as peripheral tissues. These organs respond to imminent or perceived threats that elicit a stress response, primarily culminating in the release of glucocorticoids into the systemic circulation by the adrenal glands. Although the secretion of glucocorticoids serves to protect and maintain homeostasis in the typical operation at baseline levels, inadequate regulation can lead to physiologic and psychologic pathologies. The cardiovascular system is especially susceptible to prolonged dysregulation of the HPA axis and glucocorticoid production. There is debate about whether cardiovascular health risks arise from the direct detrimental effects of stress axis activation or whether pathologies develop secondary to the accompanying metabolic strain of excess glucocorticoids. In this review, we will explore the emerging research that indicates stress does have direct effects on the cardiovascular system via the HPA axis activation, with emphasis on the latest research on the impact of glucocorticoids signaling in the vasculature and the heart.
Collapse
Affiliation(s)
- Natalie G Burford
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA 71130-3932, USA.
| | - Natalia A Webster
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA 71130-3932, USA.
| | - Diana Cruz-Topete
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA 71130-3932, USA.
| |
Collapse
|
36
|
Abstract
This article seeks to evaluate current practices in heart transplantation. The goals of this article were to review current practices for heart transplantation and its anesthesia management. The article reviews current demographics and discusses the current criteria for candidacy for heart transplantation. The process for donor and receipt selection is reviewed. This is followed by a review of mechanical circulatory support devices as they pertain to heart transplantation. The preanesthesia and intraoperative considerations are also discussed. Finally, management after transplantation is also reviewed.
Collapse
Affiliation(s)
- Davinder Ramsingh
- Department of Anesthesiology, Loma Linda Medical Center, 11234 Anderson Street, MC-2532-D, Loma Linda, CA 92354, USA.
| | - Reed Harvey
- Department of Anesthesiology, Ronald Reagan UCLA Medical Center, University of California at Los Angeles, 757 Westwood Plaza, Suite 3325, Los Angeles, CA 90095-7403, USA
| | - Alec Runyon
- Department of Anesthesiology, Loma Linda Medical Center, 11234 Anderson Street, MC-2532-D, Loma Linda, CA 92354, USA
| | - Michael Benggon
- Department of Anesthesiology, Loma Linda Medical Center, 11234 Anderson Street, MC-2532-D, Loma Linda, CA 92354, USA
| |
Collapse
|
37
|
Yaung J, Arabia FA, Nurok M. Perioperative Care of the Patient With the Total Artificial Heart. Anesth Analg 2017; 124:1412-1422. [PMID: 28107271 DOI: 10.1213/ane.0000000000001851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Advanced heart failure continues to be a leading cause of morbidity and mortality despite improvements in pharmacologic therapy. High demand for cardiac transplantation and shortage of donor organs have led to an increase in the utilization of mechanical circulatory support devices. The total artificial heart is an effective biventricular assist device that may be used as a bridge to transplant and that is being studied for destination therapy. This review discusses the history, indications, and perioperative management of the total artificial heart with emphasis on the postoperative concerns.
Collapse
Affiliation(s)
- Jill Yaung
- From the *Department of Anesthesiology, Cedars-Sinai Medical Center, Los Angeles, California; and †Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | | | | |
Collapse
|
38
|
Kachulis B, Mitrev L, Jordan D. Intraoperative anesthetic management of lung transplantation patients. Best Pract Res Clin Anaesthesiol 2017; 31:261-272. [DOI: 10.1016/j.bpa.2017.04.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 11/15/2022]
|
39
|
Khamooshian A, Hai T, Amador Y, Jeganathan J, Mahmood F, Matyal R. Intraoperative Challenges in the Management of Biventricular Failure in Takotsubo Cardiomyopathy. J Cardiothorac Vasc Anesth 2016; 31:1318-1321. [PMID: 28262450 DOI: 10.1053/j.jvca.2016.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Arash Khamooshian
- Department of Cardio-Thoracic Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Ting Hai
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology, Peking University People's Hospital, Beijing, China
| | - Yannis Amador
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesia, Hospital México, Universidad de Costa Rica, San José, Costa Rica
| | - Jelliffe Jeganathan
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Feroze Mahmood
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA.
| | - Robina Matyal
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
40
|
Łukaszyk E, Małyszko J. Copeptin: Pathophysiology and potential clinical impact. Adv Med Sci 2015; 60:335-41. [PMID: 26233637 DOI: 10.1016/j.advms.2015.07.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 06/29/2015] [Accepted: 07/02/2015] [Indexed: 12/24/2022]
Abstract
Copeptin, a C-terminal part of the precursor pre-provasopressin is a novel biomarker of arginine-vasopressin (AVP) system. Measurements of AVP concentration are not used in clinical practice because of technical difficulties. Copeptin is synthesized in stoichiometric ratio with AVP, hence it reflects vasopressin concentration in human plasma and serum. This review outlines current research concerning the role of copeptin as a prognostic marker in different diseases and its potential clinical value.
Collapse
|
41
|
Epinephrine, compared with arginine vasopressin, is associated with similar haemodynamic effects but significantly improved brain oxygenation in the early phase of anaphylactic shock in rats. Eur J Anaesthesiol 2015; 32:563-70. [DOI: 10.1097/eja.0000000000000238] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
Jafari A, Baghaei A, Solgi R, Baeeri M, Chamanara M, Hassani S, Gholami M, Ostad SN, Sharifzadeh M, Abdollahi M. An electrocardiographic, molecular and biochemical approach to explore the cardioprotective effect of vasopressin and milrinone against phosphide toxicity in rats. Food Chem Toxicol 2015; 80:182-192. [PMID: 25796571 DOI: 10.1016/j.fct.2015.02.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 02/24/2015] [Accepted: 02/25/2015] [Indexed: 12/27/2022]
Abstract
The present study was conducted to identify the protective effect of vasopressin (AVP) and milrinone on cardiovascular function, mitochondrial complex activities, cellular ATP reserve, oxidative stress, and apoptosis in rats poisoned by aluminum phosphide (AlP). Rats were divided into five groups (n = 12) including control, AlP (12.5 mg/kg), AlP + AVP (2.0 Units/kg), AlP + milrinone (0.25 mg/kg) and AlP + AVP + milrinone. After treatment, the animals were connected to an electronic cardiovascular monitoring device to monitor electrocardiographic (ECG) parameter. Finally, oxidative stress biomarkers, mitochondrial complex activities, ADP/ATP ratio and apoptosis were evaluated on the heart tissues. Results indicated that AlP administration induced ECG abnormalities along with a decline in blood pressure and heart rate. AVP and milrinone significantly ameliorated these changes in all treated groups. Considerable protective effects on oxidative stress biomarkers, complex IV activity, ADP/ATP ratio and caspase-3 and -9 activities in treated groups were also found. These findings were supported by flow cytometry assay of cardiomyocytes. In conclusion, administration of AVP and milrinone, not only improve cardiovascular functions in AlP poisoned rats in the short time, but after a long time can also restore mitochondrial function and ATP level and reduce the oxidative damage, which prevent cardiomyocytes from entering the apoptotic phase.
Collapse
Affiliation(s)
- Abbas Jafari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; and Pharmaceutical Sciences Research Center; and Poisoning & Toxicology Research Center; and Endocrinology & Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran 1417614411, Iran; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Amir Baghaei
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; and Pharmaceutical Sciences Research Center; and Poisoning & Toxicology Research Center; and Endocrinology & Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Reza Solgi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; and Pharmaceutical Sciences Research Center; and Poisoning & Toxicology Research Center; and Endocrinology & Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Maryam Baeeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; and Pharmaceutical Sciences Research Center; and Poisoning & Toxicology Research Center; and Endocrinology & Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mohsen Chamanara
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shokoufeh Hassani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; and Pharmaceutical Sciences Research Center; and Poisoning & Toxicology Research Center; and Endocrinology & Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mahdi Gholami
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; and Pharmaceutical Sciences Research Center; and Poisoning & Toxicology Research Center; and Endocrinology & Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Seyed Nasser Ostad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; and Pharmaceutical Sciences Research Center; and Poisoning & Toxicology Research Center; and Endocrinology & Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Moahmmad Sharifzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; and Pharmaceutical Sciences Research Center; and Poisoning & Toxicology Research Center; and Endocrinology & Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy; and Pharmaceutical Sciences Research Center; and Poisoning & Toxicology Research Center; and Endocrinology & Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran 1417614411, Iran.
| |
Collapse
|
43
|
Baranowska-Bik A, Kochanowski J, Uchman D, Litwiniuk A, Kalisz M, Martynska L, Wolinska-Witort E, Baranowska B, Bik W. Association of copeptin and cortisol in newly diagnosed multiple sclerosis patients. J Neuroimmunol 2015; 282:21-4. [DOI: 10.1016/j.jneuroim.2015.03.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 03/08/2015] [Accepted: 03/09/2015] [Indexed: 11/16/2022]
|
44
|
Schrimpf C, Gillmann HJ, Sahlmann B, Meinders A, Larmann J, Wilhelmi M, Aper T, Rustum S, Lichtinghagen R, Theilmeier G, Teebken OE. Renal function interferes with copeptin in prediction of major adverse cardiac events in patients undergoing vascular surgery. PLoS One 2015; 10:e0123093. [PMID: 25875814 PMCID: PMC4395325 DOI: 10.1371/journal.pone.0123093] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 02/27/2015] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE Precise perioperative risk stratification is important in vascular surgery patients who are at high risk for major adverse cardiovascular events (MACE) peri- and postoperatively. In clinical practice, the patient's perioperative risk is predicted by various indicators, e.g. revised cardiac index (RCRI) or modifications thereof. Patients suffering from chronic kidney disease (CKD) are stratified into a higher risk category. We hypothesized that Copeptin as a novel biomarker for hemodynamic stress could help to improve the prediction of perioperative cardiovascular events in patients undergoing vascular surgery including patients with chronic kidney disease. METHODS 477 consecutive patients undergoing abdominal aortic, peripheral arterial or carotid surgery from June 2007 to October 2012 were prospectively enrolled. Primary endpoint was 30-day postoperative major adverse cardiovascular events (MACE). RESULTS 41 patients reached the primary endpoint, including 63.4% aortic, 26.8% carotid, and 9.8% peripheral surgeries. Linear regression analysis showed that RCRI (P< .001), pre- (P< .001), postoperative Copeptin (P< .001) and Copeptin level change (P= .001) were associated with perioperative MACE, but CKD remained independently associated with MACE and Copeptin levels. Multivariate regression showed that increased Copeptin levels added risk predictive information to the RCRI (P= .003). Especially in the intermediate RCRI categories was Copeptin significantly associated with the occurrence of MACE. (P< .05 Kruskal Wallis test). Subdivision of the study cohort into CKD stages revealed that preoperative Copeptin was significantly associated with CKD stages (P< .0001) and preoperative Copeptin measurements could not predict MACE in patients with more severe CKD stages. CONCLUSION Preoperative Copeptin loses its risk predictive potential for perioperative MACE in patients with chronic kidney disease undergoing vascular surgery.
Collapse
Affiliation(s)
- Claudia Schrimpf
- Division of Vascular & Endovascular Surgery, Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Hans-Joerg Gillmann
- Department for Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Bianca Sahlmann
- Department for Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Antje Meinders
- Department for Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Jan Larmann
- Department for Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Mathias Wilhelmi
- Division of Vascular & Endovascular Surgery, Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Thomas Aper
- Division of Vascular & Endovascular Surgery, Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Saad Rustum
- Division of Vascular & Endovascular Surgery, Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Ralf Lichtinghagen
- Department for Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Gregor Theilmeier
- Department for Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Omke E. Teebken
- Division of Vascular & Endovascular Surgery, Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
45
|
Then C, Kowall B, Lechner A, Meisinger C, Heier M, Koenig W, Peters A, Rathmann W, Seissler J. Plasma copeptin is associated with type 2 diabetes in men but not in women in the population-based KORA F4 study. Acta Diabetol 2015; 52:103-12. [PMID: 25011650 DOI: 10.1007/s00592-014-0609-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 05/29/2014] [Indexed: 01/12/2023]
Abstract
Elevated plasma CT-pro-vasopressin (copeptin) has been described as biomarkers for type 2 diabetes (T2D) and the metabolic syndrome (MetS), which, however, was not confirmed by all studies. Here, we analyzed the association of copeptin with T2D, MetS and MetS components in the population-based KORA F4 study. Plasma copeptin concentrations were analyzed in 1,554 study participants. We used fractional polynomial selection procedures to check for nonlinearity of the associations between copeptin and T2D and HbA1c, respectively. In logistic regression models, we investigated associations between copeptin and T2D, MetS and its components according to IDF criteria. In the fractional polynomial approach, linear models fitted best for copeptin. In multivariable adjusted models, copeptin as a continuous variable was associated with T2D and HbA1c only in men (OR = 1.38 per standard deviation, 95 % CI 1.13-1.70 for T2D). Comparing the top quartile Q4 versus Q1-3, elevated copeptin was associated with T2D (OR 2.70, 95 % CI 1.60-4.59) in men but not in women (OR 0.98, 95 % CI 0.52-1.83). Copeptin was not significantly associated with MetS, central obesity, triglycerides and reduced HDL cholesterol. A significant association with copeptin was observed for hypertension in women (OR 1.59, 95 % CI 1.08-2.33) and glucose dysfunction according to IDF criteria in men (OR 1.63, 95 % CI 1.14-2.34). In the KORA F4 study, copeptin was significantly associated with T2D only in men, whereas hypertension was associated with copeptin in women. No other components of the MetS were related to elevated copeptin.
Collapse
Affiliation(s)
- Cornelia Then
- Medizinische Klinik und Poliklinik IV, Diabetes Zentrum, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Ziemssenstraße 1, 80336, Munich, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dal Lin C, Tona F, Osto E. Coronary Microvascular Function and Beyond: The Crosstalk between Hormones, Cytokines, and Neurotransmitters. Int J Endocrinol 2015; 2015:312848. [PMID: 26124827 PMCID: PMC4466475 DOI: 10.1155/2015/312848] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/10/2015] [Accepted: 03/16/2015] [Indexed: 01/18/2023] Open
Abstract
Beyond its hemodynamic function, the heart also acts as a neuroendocrine and immunoregulatory organ. A dynamic communication between the heart and other organs takes place constantly to maintain cardiovascular homeostasis. The current understanding highlights the importance of the endocrine, immune, and nervous factors to fine-tune the crosstalk of the cardiovascular system with the entire body. Once disrupted, this complex interorgan communication may promote the onset and the progression of cardiovascular diseases. Thus, expanding our knowledge on how these factors influence the cardiovascular system can lead to novel therapeutic strategies to improve patient care. In the present paper, we review novel concepts on the role of endocrine, immune, and nervous factors in the modulation of microvascular coronary function.
Collapse
Affiliation(s)
- Carlo Dal Lin
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Via Giustiniani 2, 35100 Padua, Italy
| | - Francesco Tona
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Via Giustiniani 2, 35100 Padua, Italy
| | - Elena Osto
- Centre for Molecular Cardiology, University of Zurich and University Heart Center, Department of Cardiology, University Hospital, Raemistrasse 100, 8091 Zurich, Switzerland
- *Elena Osto:
| |
Collapse
|
47
|
Pathophysiology of Portal Hypertension. PANVASCULAR MEDICINE 2015. [PMCID: PMC7153457 DOI: 10.1007/978-3-642-37078-6_144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The bases of our current knowledge on the physiology of the hepatic portal system are largely owed to the work of three pioneering vascular researchers from the sixteenth and the seventeenth centuries: A. Vesalius, W. Harvey, and F. Glisson. Vesalius is referred to as the founder of modern human anatomy, and in his influential book, De humani corporis fabrica libri septem, he elaborated the first anatomical atlas of the hepatic portal venous system (Vesalius 2013). Sir William Harvey laid the foundations of modern cardiovascular research with his Exercitatio Anatomica de Motu Cordis et Sanguinis in Animalibus (Harvey 1931) in which he established the nature of blood circulation. Finally, F. Glisson characterized the gastrointestinal-hepatic vascular system (Child 1955). These physiological descriptions were later complemented with clinical observations. In the eighteenth and nineteenth centuries, Morgagni, Puckelt, Cruveilhier, and Osler were the first to make the connection between common hepatic complications – ascites, splenomegaly, and gastrointestinal bleeding – and obstruction of the portal system (Sandblom 1993). These were the foundations that allowed Gilbert, Villaret, and Thompson to establish an early definition of portal hypertension at the beginning of the twentieth century. In this period, Thompson performed the first direct measurement of portal pressure by laparotomy in some patients (Gilbert and Villaret 1906; Thompson et al. 1937). Considering all these milestones, and paraphrasing Sir Isaac Newton, if hepatologists have seen further, it is by standing on the shoulders of giants. Nowadays, our understanding of the pathogenesis of portal hypertension has largely improved thanks to the progress in preclinical and clinical research. However, this field is ever-changing and hepatologists are continually identifying novel pathological mechanisms and developing new therapeutic strategies for this clinical condition. Hence, the aim of this chapter is to summarize the current knowledge about this clinical condition.
Collapse
|
48
|
Zimmermann-Peruzatto JM, Lazzari VM, de Moura AC, Almeida S, Giovenardi M. Examining the Role of Vasopressin in the Modulation of Parental and Sexual Behaviors. Front Psychiatry 2015; 6:130. [PMID: 26441691 PMCID: PMC4585274 DOI: 10.3389/fpsyt.2015.00130] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 09/04/2015] [Indexed: 11/13/2022] Open
Abstract
Vasopressin (VP) and VP-like neuropeptides are evolutionarily stable peptides found in all vertebrate species. In non-mammalian vertebrates, vasotocin (VT) plays a role similar to mammalian VP, whereas mesotocin and isotocin are functionally similar to mammalian oxytocin (OT). Here, we review the involvement of VP in brain circuits, synaptic plasticity, evolution, and function, highlighting the role of VP in social behavior. In all studied species, VP is encoded on chromosome 20p13, and in mammals, VP is produced in specific hypothalamic nuclei and released by the posterior pituitary. The role of VP is mediated by the stimulation of the V1a, V1b, and V2 receptors as well as the oxytocinergic and purinergic receptors. VT and VP functions are usually related to osmotic and cardiovascular homeostasis when acting peripherally. However, these neuropeptides are also critically involved in the central modulation of social behavior displays, such as pairing recognition, pair-bonding, social memory, sexual behavior, parental care, and maternal and aggressive behavior. Evidence suggests that these effects are primarily mediated by V1a receptor in specific brain circuits that provide important information for the onset and control of social behaviors in normal and pathological conditions.
Collapse
Affiliation(s)
- Josi Maria Zimmermann-Peruzatto
- Programa de Pós-Graduação em Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS) , Porto Alegre , Brazil
| | - Virgínia Meneghini Lazzari
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA) , Porto Alegre , Brazil
| | - Ana Carolina de Moura
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA) , Porto Alegre , Brazil
| | - Silvana Almeida
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA) , Porto Alegre , Brazil
| | - Márcia Giovenardi
- Programa de Pós-Graduação em Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS) , Porto Alegre , Brazil ; Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA) , Porto Alegre , Brazil
| |
Collapse
|
49
|
Martin DT, Schreiber MA. Modern resuscitation of hemorrhagic shock: what is on the horizon? Eur J Trauma Emerg Surg 2014; 40:641-656. [PMID: 26814779 DOI: 10.1007/s00068-014-0416-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 05/23/2014] [Indexed: 12/24/2022]
Abstract
PURPOSE Mortality rates among the severely injured remain high. The successful treatment of hemorrhagic shock relies on expeditious control of bleeding through surgical ligation, packing, or endovascular techniques. An important secondary concern in hemorrhaging patients is how to respond to the lost blood volume. A single method that is able to adequately address all needs of the exsanguinating patient has not yet been agreed upon, despite a large growth of knowledge regarding the causative factors of traumatic shock. METHODS A review of relevent literature was performed. CONCLUSIONS Many different trials are currently underway to discriminate ways to improve outcomes in the severely injured and bleeding patient. This paper will review: (1) recent advances in our understanding of the effects hemorrhagic shock has on the coagulation cascade and vascular endothelium, (2) recent research findings that have changed resuscitation, and (3) resuscitation strategies that are not widely used but under active investigation.
Collapse
Affiliation(s)
- D T Martin
- Division of Trauma, Critical Care, and Acute Care Surgery, Department of Surgery, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L-611, Portland, OR, 97239, USA.
- Division of Trauma, Critical Care, and Acute Care Surgery, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L-611, Portland, OR, 97239, USA.
| | - M A Schreiber
- Division of Trauma, Critical Care, and Acute Care Surgery, Department of Surgery, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L-611, Portland, OR, 97239, USA.
- Division of Trauma, Critical Care, and Acute Care Surgery, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L-611, Portland, OR, 97239, USA.
| |
Collapse
|
50
|
Sherwi N, Pellicori P, Joseph AC, Buga L. Old and newer biomarkers in heart failure: from pathophysiology to clinical significance. J Cardiovasc Med (Hagerstown) 2014; 14:690-7. [PMID: 23846675 DOI: 10.2459/jcm.0b013e328361d1ef] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Heart failure is a complex disease in which a careful clinical examination and the measurement of cardiac function may not always be sufficient for making a correct diagnosis. Measuring plasma levels of natriuretic peptides may assist in this process, also offering a good tool for accurate risk stratification. Other alternative biomarkers may give insight into the different pathways of heart failure genesis and pathophysiology, and may help to identify those patients with overt heart failure and a more adverse outcome, or distinguish between those at risk of developing heart failure. Despite a high number of potentially useful biomarkers, only a few will likely be introduced routinely into clinical practice. However, a multi-marker approach might increase the diagnostic accuracy and it might identify different phenotypes of heart failure patients who might benefit from individualized therapy in the future.
Collapse
Affiliation(s)
- Nasser Sherwi
- Department of Academic Cardiology, Hull and East Yorkshire Medical Research and Teaching Centre, Castle Hill Hospital, Cottingham, Kingston upon Hull, UK
| | | | | | | |
Collapse
|