151
|
Lehtimäki J, Ventura-Aquino E, Chu X, Paredes RG, Ågmo A. Sexual Incentive Motivation and Copulatory Behavior in Male Rats Treated With the Adrenergic α 2-Adrenoceptor Agonists Tasipimidine and Fadolmidine: Implications for Treatment of Premature Ejaculation. J Sex Med 2021; 18:1677-1689. [PMID: 37057503 DOI: 10.1016/j.jsxm.2021.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 07/06/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Premature ejaculation is the most common sexual dysfunction in young men, and it often leads to reduced relationship satisfaction and quality of life. AIM To determine the role of central and peripheral α2-adrenoceptors in the control of ejaculation and sexual incentive motivation in rats. METHODS Sexual incentive motivation was studied in a large arena in which a male subject could choose between approaching and remaining close to a sexually receptive female or another male. Sexual behavior was studied in standard observation cages in which a male was allowed to freely interact with a receptive female for 30 minutes. Two highly selective agonists at the α2-adrenoceptors, tasipimidine and fadolmidine, were administered before the tests. Low peripheral doses of fadolmidine have been reported to have effects mainly outside of the central nervous system, whereas at large doses also the central effects are evident. OUTCOMES The time spent close to the receptive female in relation to the time spent with the male and measures of ambulatory activity were obtained from the test for sexual incentive motivation, while the habitual parameters of sexual behavior were recorded with the copulation test. RESULTS Tasipimidine prolonged ejaculation latency and the interintromission interval at the dose of 200 µg/kg when data from fast-ejaculating rats were used. No other sexual parameter was modified. A dose of 100 µg/kg was ineffective. There was no consistent effect on sexual incentive motivation, although modest sedation was observed. Fadolmidine, a drug that does not easily penetrate the blood-brain barrier, had no effect on sexual incentive motivation at any of the doses used (3, 30, and 100 µg/kg). The largest dose had clear sedative effects. The lower doses had no systematic effect on sexual behavior, not even when only fast or very fast ejaculating males were analyzed. CLINICAL TRANSLATION The findings are relevant to the search for treatments for premature ejaculation that are specific enough to selectively delay ejaculation. STRENGTHS & LIMITATIONS The procedures used here are standard in the field and yield the most reliable data. Whether the effects observed in male rats are directly transferrable to men can only be determined through clinical studies. CONCLUSION The observation that drugs acting at central but not peripheral α2-adrenoceptors prolong ejaculation latency without affecting any other parameter of sexual behavior or sexual incentive motivation suggests that this kind of drug may be suitable for treating premature ejaculation. Jyrki L., Elisa V.-A., Xi C., et al. Sexual Incentive Motivation and Copulatory Behavior in Male Rats Treated With the Adrenergic α2-Adrenoceptor Agonists Tasipimidine and Fadolmidine: Implications for Treatment of Premature Ejaculation. J Sex Med 2021;18:1677-1689.
Collapse
Affiliation(s)
| | - Elisa Ventura-Aquino
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla UNAM, Querétaro, México
| | - Xi Chu
- Department of Psychology, NTNU, Trondheim, Norway; Department of Psychology, University of Tromsø, Tromsø, Norway
| | - Raúl G Paredes
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla UNAM, Querétaro, México; Instituto de Neurobiología, UNAM Querétaro, México
| | - Anders Ågmo
- Department of Psychology, University of Tromsø, Tromsø, Norway
| |
Collapse
|
152
|
A mononuclear PdII complex with Naphcon; crystal structure, experimental and computational studies of the interaction with DNA/BSA and evaluation of anticancer activity. Polyhedron 2021. [DOI: 10.1016/j.poly.2021.115333] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
153
|
Kim YS, Chang HW, Kim H, Park JS, Won YJ. Comparison of the effects of dexmedetomidine and remifentanil on perioperative hemodynamics and recovery profile of patients undergoing laryngeal microsurgery: A prospective randomized double-blinded study. Medicine (Baltimore) 2021; 100:e27045. [PMID: 34449490 PMCID: PMC8389956 DOI: 10.1097/md.0000000000027045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/03/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Laryngeal microsurgery (LMS) causes hemodynamic instability and postoperative agitation, cough, pain, nausea, and vomiting. Moreover, because of a short operation time, it is associated with challenging anesthetic management. The aim of this study was to compare the usefulness of continuous administration of dexmedetomidine and remifentanil in inducing general anesthesia in patients undergoing LMS. METHODS This is a prospective randomized control design. Continuous intravenous infusion of dexmedetomidine (group D) or remifentanil (group R) was administered from 10 minutes before the induction of anesthesia to the end of surgery. In both groups, 1.5 mg/kg propofol and 0.5 mg/kg rocuronium were administered for the induction of anesthesia, and desflurane were titrated during the measurement of the bispectral index. We recorded hemodynamic data, recovery time, grade of cough, pain score, and analgesic requirements during the perioperative period. RESULTS 61 patients were finally analyzed (30 for group D, 31 for group R). The incidence of moderate to severe postoperative sore throat was higher in group R than in group D (42% vs 10%, P = .008), and the quantity of rescue fentanyl used in post-anesthesia care unit was significantly higher in group R than in group D (23.2 ± 24.7 mg vs 3.3 ± 8.6 mg; P < .001); however, the time required for eye opening was significantly longer in group D than in group R (599.4 ± 177.9 seconds vs 493.5 ± 103.6 seconds; P = .006). The proportion of patients with no cough or single cough during extubation was comparable between the 2 groups (group D vs group R: 73% vs 70%) as was the incidence of hemodynamic instability. CONCLUSION Although there was a transient delay in emergence time, dexmedetomidine reduced postoperative opioid use and the incidence of sore throat. Dexmedetomidine may be used as an alternative agent to opioids in patients undergoing LMS.
Collapse
Affiliation(s)
- Young Sung Kim
- Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hae Wone Chang
- Department of Anesthesiology and Pain Medicine, Eulji University Hospital, Seoul, Republic of Korea
| | - Heezoo Kim
- Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jong Sun Park
- Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Young Ju Won
- Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
154
|
Arbabi A, Bao X, Shalaby WS, Razeghinejad R. Systemic side effects of glaucoma medications. Clin Exp Optom 2021; 105:157-165. [PMID: 34402741 DOI: 10.1080/08164622.2021.1964331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Glaucoma is a progressive loss of retinal ganglion cells leading to visual field loss. Lowering intraocular pressure is currently the only modifiable risk factor to slow glaucoma progression. Intraocular pressure-lowering options include topical and systemic medications, lasers, and surgical procedures. Glaucoma eye drops play a major role in treating this blinding disease. Similar to all medications, the glaucoma medications have their own adverse effects. The majority of glaucoma medications work by stimulating or inhibiting adrenergic, cholinergic, and prostaglandin receptors, which are distributed all over the body. Therefore, the glaucoma medications can affect organs other than the eye. This review will discuss the systemic adverse effects of carbonic anhydrase inhibitors, sympathomimetics, para-sympathomimetics, beta blockers, prostaglandin analogs, hyperosmotic agents, and novel glaucoma medications with a stress on pregnant patients, breastfeeding mothers, and paediatric patients.
Collapse
Affiliation(s)
- Amirmohsen Arbabi
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, Pennsylvania, USA
| | - Xuan Bao
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
| | - Wesam Shamseldin Shalaby
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, Pennsylvania, USA.,Tanta Medical School, Tanta University, Tanta, Gharbia, Egypt
| | - Reza Razeghinejad
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, Pennsylvania, USA
| |
Collapse
|
155
|
Elmarakby A, Sullivan J. Sex differences in hypertension: lessons from spontaneously hypertensive rats (SHR). Clin Sci (Lond) 2021; 135:1791-1804. [PMID: 34338771 PMCID: PMC8329852 DOI: 10.1042/cs20201017] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/30/2022]
Abstract
Although numerous clinical and experimental studies have clearly identified a sexual dimorphism in blood pressure control, the mechanism(s) underlying gender differences in blood pressure remain unclear. Over the past two decades, numerous laboratories have utilized the spontaneously hypertensive rats (SHR) as an experimental model of essential hypertension to increase our understanding of the mechanisms regulating blood pressure in males and females. Previous work by our group and others have implicated that differential regulation of adrenergic receptors, the renin-angiotensin system, oxidative stress, nitric oxide bioavailability and immune cells contribute to sex differences in blood pressure control in SHR. The purpose of this review is to summarize previous findings to date regarding the mechanisms of blood pressure control in male versus female SHR.
Collapse
Affiliation(s)
- Ahmed A. Elmarakby
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA 30912, U.S.A
| | | |
Collapse
|
156
|
Jimenez-Tellez N, Iqbal F, Pehar M, Casas-Ortiz A, Rice T, Syed NI. Dexmedetomidine does not compromise neuronal viability, synaptic connectivity, learning and memory in a rodent model. Sci Rep 2021; 11:16153. [PMID: 34373548 PMCID: PMC8352930 DOI: 10.1038/s41598-021-95635-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/21/2021] [Indexed: 02/08/2023] Open
Abstract
Recent animal studies have drawn concerns regarding most commonly used anesthetics and their long-term cytotoxic effects, specifically on the nervous tissue. It is therefore imperative that the search continues for agents that are non-toxic at both the cellular and behavioural level. One such agent appears to be dexmedetomidine (DEX) which has not only been found to be less neurotoxic but has also been shown to protect neurons from cytotoxicity induced by other anesthetic agents. However, DEX's effects on the growth and synaptic connectivity at the individual neuronal level, and the underlying mechanisms have not yet been fully resolved. Here, we tested DEX for its impact on neuronal growth, synapse formation (in vitro) and learning and memory in a rodent model. Rat cortical neurons were exposed to a range of clinically relevant DEX concentrations (0.05-10 µM) and cellular viability, neurite outgrowth, synaptic assembly and mitochondrial morphology were assessed. We discovered that DEX did not affect neuronal viability when used below 10 µM, whereas significant cell death was noted at higher concentrations. Interestingly, in the presence of DEX, neurons exhibited more neurite branching, albeit with no differences in corresponding synaptic puncta formation. When rat pups were injected subcutaneously with DEX 25 µg/kg on postnatal day 7 and again on postnatal day 8, we discovered that this agent did not affect hippocampal-dependent memory in freely behaving animals. Our data demonstrates, for the first time, the non-neurotoxic nature of DEX both in vitro and in vivo in an animal model providing support for its utility as a safer anesthetic agent. Moreover, this study provides the first direct evidence that although DEX is growth permissive, causes mitochondrial fusion and reduces oxygen reactive species production, it does not affect the total number of synaptic connections between the cortical neurons in vitro.
Collapse
Affiliation(s)
- Nerea Jimenez-Tellez
- grid.22072.350000 0004 1936 7697Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada ,grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, Calgary, Canada ,grid.413571.50000 0001 0684 7358Alberta Children’s Hospital Research Institute, Calgary, Canada
| | - Fahad Iqbal
- grid.413571.50000 0001 0684 7358Alberta Children’s Hospital Research Institute, Calgary, Canada
| | - Marcus Pehar
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, Calgary, Canada ,grid.413571.50000 0001 0684 7358Alberta Children’s Hospital Research Institute, Calgary, Canada
| | - Alberto Casas-Ortiz
- grid.22072.350000 0004 1936 7697Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada ,grid.413571.50000 0001 0684 7358Alberta Children’s Hospital Research Institute, Calgary, Canada
| | - Tiffany Rice
- grid.413571.50000 0001 0684 7358Alberta Children’s Hospital Research Institute, Calgary, Canada ,grid.22072.350000 0004 1936 7697Department of Anesthesiology, Perioperative and Pain Medicine, University of Calgary, Calgary, Canada
| | - Naweed I. Syed
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, Calgary, Canada ,grid.413571.50000 0001 0684 7358Alberta Children’s Hospital Research Institute, Calgary, Canada ,grid.22072.350000 0004 1936 7697Department of Cell Biology and Anatomy, University of Calgary, Calgary, Canada
| |
Collapse
|
157
|
Leino T, Viitamaa T, Salonen JS, Pesonen U, Haapalinna A. Effects of fadolmidine, an α 2 -adrenoceptor agonist, as an adjuvant to spinal bupivacaine on antinociception and motor function in rats and dogs. Pharmacol Res Perspect 2021; 9:e00830. [PMID: 34302721 PMCID: PMC8308519 DOI: 10.1002/prp2.830] [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: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 11/06/2022] Open
Abstract
α2 -Adrenoceptor agonists such as clonidine and dexmedetomidine are used as adjuvants to local anesthetics in regional anesthesia. Fadolmidine is an α2 -adrenoceptor agonist developed especially as a spinal analgesic. The current studies investigate the effects of intrathecally administered fadolmidine with a local anesthetic, bupivacaine, on antinociception and motor block in conscious rats and dogs. The antinociceptive effects of intrathecal fadolmidine and bupivacaine alone or in combination were tested in the rat tail-flick and the dog's skin twitch models. The durations of motor block in rats and in dogs were also assessed. In addition, the effects on sedation, mean arterial blood pressure, heart rate, respiratory rate and body temperature were evaluated in telemetrized dogs. Concentrations of fadolmidine in plasma and spinal cord were determined after intrathecal and intravenous administration in rats. Co-administration of intrathecal fadolmidine with bupivacaine increased the magnitude and duration of the antinociceptive effects and prolonged motor block without hypotension. The interaction of the antinociceptive effect was synergistic in its nature in rats. Concentration of fadolmidine in plasma was very low after intrathecal dosing. Taken together, these studies show that fadolmidine as an adjuvant to intrathecal bupivacaine provides enhanced sensory-motor block and enables a reduction of the doses of both drugs. The results indicate that co-administration of fadolmidine with intrathecal bupivacaine was able to achieve an enhanced antinociceptive effect without hypotension and could thus represent a suitable combination for spinal anesthesia.
Collapse
Affiliation(s)
- Tiina Leino
- Orion Corporation Orion PharmaR&DTurkuFinland
| | | | | | - Ullamari Pesonen
- Integrative Physiology and Pharmacology Research UnitInstitute of BiomedicineFaculty of MedicineUniversity of TurkuTurkuFinland
| | | |
Collapse
|
158
|
Miller DL, Dou C, Raghavendran K, Dong Z. The Influence of Xylazine and Clonidine on Lung Ultrasound-Induced Pulmonary Capillary Hemorrhage in Spontaneously Hypertensive Rats. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:2331-2338. [PMID: 33972153 PMCID: PMC8243848 DOI: 10.1016/j.ultrasmedbio.2021.03.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/18/2021] [Accepted: 03/29/2021] [Indexed: 05/21/2023]
Abstract
Induction of pulmonary capillary hemorrhage (PCH) by lung ultrasound (LUS) depends not only on physical exposure parameters but also on physiologic conditions and drug treatment. We studied the influence of xylazine and clonidine on LUS-induced PCH in spontaneously hypertensive and normotensive rats using diagnostic B-mode ultrasound at 7.3 MHz. Using ketamine anesthesia, rats receiving saline, xylazine, or clonidine treatment were tested with different pulse peak rarefactional pressure amplitudes in 5 min exposures. Results with xylazine or clonidine in spontaneously hypertensive rats were not significantly different at the three exposure pulse peak rarefactional pressure amplitudes, and thresholds were lower (2.2 MPa) than with saline (2.6 MPa). Variations in LUS PCH were not correlated with mean systemic blood pressure. Similar to previous findings for dexmedetomidine, the clinical drug clonidine tended to increase susceptibility to LUS PCH.
Collapse
Affiliation(s)
- Douglas L Miller
- Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan, USA.
| | - Chunyan Dou
- Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Krishnan Raghavendran
- Department of Surgery, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Zhihong Dong
- Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan, USA
| |
Collapse
|
159
|
Dominguini D, Steckert AV, Michels M, Spies MB, Ritter C, Barichello T, Thompson J, Dal-Pizzol F. The effects of anaesthetics and sedatives on brain inflammation. Neurosci Biobehav Rev 2021; 127:504-513. [PMID: 33992694 DOI: 10.1016/j.neubiorev.2021.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 04/27/2021] [Accepted: 05/09/2021] [Indexed: 12/17/2022]
Abstract
Microglia are involved in many dynamic processes in the central nervous system (CNS) including the development of inflammatory processes and neuromodulation. Several sedative, analgesic or anaesthetic drugs, such as opioids, ∝2-adrenergic agonists, ketamine, benzodiazepines and propofol can cause both neuroprotective and harmful effects on the brain. The purpose of this review is to present the main findings on the use of these drugs and the mechanisms involved in microglial activation. Alpha 2-adrenergic agonists, propofol and benzodiazepines have several pro- or anti-inflammatory effects on microglia. Long-term use of benzodiazepines and propofol causes neuroapoptotic effects and α2-adrenergic agonists may attenuate these effects. Conversely, morphine and fentanyl may have proinflammatory effects, causing behavioural changes in patients and changes in cell viability in vitro. Conversely, chronic administration of morphine induces CCL5 chemokine expression in microglial cells that promotes their survival.
Collapse
Affiliation(s)
- Diogo Dominguini
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Amanda V Steckert
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Monique Michels
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Mariana B Spies
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Cristiane Ritter
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Jonathan Thompson
- Department of Cardiovascular Sciences, Anaesthesia Critical Care and Pain Management Group, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, UK
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
160
|
Archer M, Dogra N, Dovey Z, Ganta T, Jang HS, Khusid JA, Lantz A, Mihalopoulos M, Stockert JA, Zahalka A, Björnebo L, Gaglani S, Noh MR, Kaplan SA, Mehrazin R, Badani KK, Wiklund P, Tsao K, Lundon DJ, Mohamed N, Lucien F, Padanilam B, Gupta M, Tewari AK, Kyprianou N. Role of α- and β-adrenergic signaling in phenotypic targeting: significance in benign and malignant urologic disease. Cell Commun Signal 2021; 19:78. [PMID: 34284799 PMCID: PMC8290582 DOI: 10.1186/s12964-021-00755-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/28/2021] [Indexed: 01/17/2023] Open
Abstract
The urinary tract is highly innervated by autonomic nerves which are essential in urinary tract development, the production of growth factors, and the control of homeostasis. These neural signals may become dysregulated in several genitourinary (GU) disease states, both benign and malignant. Accordingly, the autonomic nervous system is a therapeutic target for several genitourinary pathologies including cancer, voiding dysfunction, and obstructing nephrolithiasis. Adrenergic receptors (adrenoceptors) are G-Protein coupled-receptors that are distributed throughout the body. The major function of α1-adrenoceptors is signaling smooth muscle contractions through GPCR and intracellular calcium influx. Pharmacologic intervention of α-and β-adrenoceptors is routinely and successfully implemented in the treatment of benign urologic illnesses, through the use of α-adrenoceptor antagonists. Furthermore, cell-based evidence recently established the antitumor effect of α1-adrenoceptor antagonists in prostate, bladder and renal tumors by reducing neovascularity and impairing growth within the tumor microenvironment via regulation of the phenotypic epithelial-mesenchymal transition (EMT). There has been a significant focus on repurposing the routinely used, Food and Drug Administration-approved α1-adrenoceptor antagonists to inhibit GU tumor growth and angiogenesis in patients with advanced prostate, bladder, and renal cancer. In this review we discuss the current evidence on (a) the signaling events of the autonomic nervous system mediated by its cognate α- and β-adrenoceptors in regulating the phenotypic landscape (EMT) of genitourinary organs; and (b) the therapeutic significance of targeting this signaling pathway in benign and malignant urologic disease. Video abstract.
Collapse
Affiliation(s)
- M. Archer
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - N. Dogra
- Department of Pathology and Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Z. Dovey
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - T. Ganta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY USA
| | - H.-S. Jang
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - J. A. Khusid
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. Lantz
- Department of Molecular Medicine and Surgery, Section of Urology, Karolinska Institute, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - M. Mihalopoulos
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - J. A. Stockert
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. Zahalka
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - L. Björnebo
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - S. Gaglani
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - M. R. Noh
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - S. A. Kaplan
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - R. Mehrazin
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - K. K. Badani
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - P. Wiklund
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - K. Tsao
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY USA
| | - D. J. Lundon
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - N. Mohamed
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - F. Lucien
- Department of Urology, Mayo Clinic, Rochester, MN USA
| | - B. Padanilam
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - M. Gupta
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. K. Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - N. Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Department of Pathology and Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| |
Collapse
|
161
|
Hassan R, Sreenivasan S, Müller CP, Hassan Z. Methadone, Buprenorphine, and Clonidine Attenuate Mitragynine Withdrawal in Rats. Front Pharmacol 2021; 12:708019. [PMID: 34322028 PMCID: PMC8311127 DOI: 10.3389/fphar.2021.708019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Kratom or Mitragyna speciosa Korth has been widely used to relieve the severity of opioid withdrawal in natural settings. However, several studies have reported that kratom may by itself cause dependence following chronic consumption. Yet, there is currently no formal treatment for kratom dependence. Mitragynine, is the major psychoactive alkaloid in kratom. Chronic mitragynine treatment can cause addiction-like symptoms in rodent models including withdrawal behaviour. In this study we assessed whether the prescription drugs, methadone, buprenorphine and clonidine, could mitigate mitragynine withdrawal effects. In order to assess treatment safety, we also evaluated hematological, biochemical and histopathological treatment effects. Methods: We induced mitragynine withdrawal behaviour in a chronic treatment paradigm in rats. Methadone (1.0 mg/kg), buprenorphine (0.8 mg/kg) and clonidine (0.1 mg/kg) were i.p. administered over four days during mitragynine withdrawal. These treatments were stopped and withdrawal sign assessment continued. Thereafter, toxicological profiles of the treatments were evaluated in the blood and in organs. Results: Chronic mitragynine treatment caused significant withdrawal behaviour lasting at least 5 days. Methadone, buprenorphine, as well as clonidine treatments significantly attenuated these withdrawal signs. No major effects on blood or organ toxicity were observed. Conclusion: These data suggest that the already available prescription medications methadone, buprenorphine, and clonidine are capable to alleviate mitragynine withdrawal signs rats. This may suggest them as treatment options also for problematic mitragynine/kratom use in humans.
Collapse
Affiliation(s)
- Rahimah Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Minden, Malaysia
| | - Sasidharan Sreenivasan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden, Malaysia
| | - Christian P Müller
- Centre for Drug Research, Universiti Sains Malaysia, Minden, Malaysia.,Section of Addiction Medicine, Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Minden, Malaysia.,Addiction Behaviour and Neuroplasticity Laboratory, National Neuroscience Institute, Singapore, Singapore
| |
Collapse
|
162
|
Wu Y, Zeng L, Zhao S. Ligands of Adrenergic Receptors: A Structural Point of View. Biomolecules 2021; 11:936. [PMID: 34202543 PMCID: PMC8301793 DOI: 10.3390/biom11070936] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/09/2021] [Accepted: 06/12/2021] [Indexed: 01/14/2023] Open
Abstract
Adrenergic receptors are G protein-coupled receptors for epinephrine and norepinephrine. They are targets of many drugs for various conditions, including treatment of hypertension, hypotension, and asthma. Adrenergic receptors are intensively studied in structural biology, displayed for binding poses of different types of ligands. Here, we summarized molecular mechanisms of ligand recognition and receptor activation exhibited by structure. We also reviewed recent advances in structure-based ligand discovery against adrenergic receptors.
Collapse
Affiliation(s)
- Yiran Wu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; (Y.W.); (L.Z.)
| | - Liting Zeng
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; (Y.W.); (L.Z.)
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; (Y.W.); (L.Z.)
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
163
|
Jiang J, Shen H, Zhang J, Wu Z, Shao X, Cui J, Zhang B, Ma X. Comparative Study of the Adverse Events Associated With Adjuvant Use of Dexmedetomidine and Clonidine in Local Anesthesia. Front Med (Lausanne) 2021; 8:602966. [PMID: 34249955 PMCID: PMC8264190 DOI: 10.3389/fmed.2021.602966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Although clonidine and dexmedetomidine are used as alpha-2 agonists to improve the quality and duration of blockade induced by local anesthetics, no study has been reported to compare their associated adverse events in local anesthesia. The aim of this study is to compare the adverse events associated with the adjuvant use of dexmedetomidine and clonidine in local anesthesia. Methods: A comprehensive search was performed to retrieve any reported adverse event associated with adjuvant use of dexmedetomidine and clonidine in local anesthesia from published literature up to 1 July 2020. Assessment of the quality of included studies was performed by the Jadad score. A comparison of any reported adverse event was made between interventions by pooling data from studies using a direct meta-analysis technique. Dichotomous outcomes were summarized as risk ratios. The review was performed according to PRISMA guideline. Results: From 121 articles retrieved from the search finally 14 articles including 1,120 patients had eligibility criteria for including in the meta-analysis. No significant difference was observed between bradycardia/hypotension (OR = 1.17; 95 % CI = 0.66–2.10; P = 0.580; I2 = 53.78 %, P = 0.027), nausea/vomiting (OR = 0.91; 95% CI = 0.59-1.42; P = 0.706; I2 = 0.0 %, P = 0.940) dizziness/headache (OR = 1.10; 95% CI = 0.44–2.75; P = 0.831; I2 = 0.0 %, P = 0.882) shivering (OR = 0.95 % CI = 0.50–1.66; P = 0.831; I2 = 0.0 %, P = 0.920) and dry mouth (OR = 1.00; 95 % CI = 0.50–1.96; P = 0.996; I2 = 0.0%, P = 0.900). No significant difference was observed in subgroup comparison of adverse events in the intravenous or local adjuvant use of the study drugs (p > 0.05). Conclusion: There is no difference in adverse events associated with the intravenous or local adjuvant use of dexmedetomidine and clonidine in local anesthesia.
Collapse
Affiliation(s)
- Jinjin Jiang
- Department of Anesthesiology, Shaoxing Shangyu Maternal and Child Health Hospital, Shaoxing, China
| | - Huasu Shen
- Department of Anesthesiology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, China
| | - Jin Zhang
- Department of Anesthesiology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, China
| | - Zhen Wu
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xian Shao
- Department of Anesthesiology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, China
| | - Jingjing Cui
- Department of Anesthesiology, Cangzhou Hospital of Integrated TCM-WM Hebei, Cangzhou, China
| | - Bao Zhang
- Department of Anesthesiology, Cangzhou Hospital of Integrated TCM-WM Hebei, Cangzhou, China
| | - Xiaoyu Ma
- Department of Anesthesiology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, China
| |
Collapse
|
164
|
Mechler K, Banaschewski T, Hohmann S, Häge A. Evidence-based pharmacological treatment options for ADHD in children and adolescents. Pharmacol Ther 2021; 230:107940. [PMID: 34174276 DOI: 10.1016/j.pharmthera.2021.107940] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/06/2021] [Indexed: 12/15/2022]
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder characterized by inattention, hyperactivity, and impulsivity, causing functional impairment. Its prevalence lies at approximately 5% in children and adolescents and at approximately 2.5% in adults. The disorder follows a multifactorial etiology and shows a high heritability. Patients show a high interindividual and intraindividual variability of symptoms, with executive deficits in several cognitive domains. Overall, ADHD is associated with high rates of psychiatric comorbidities, and insufficient treatment is linked to adverse long-term outcomes. Current clinical guidelines recommend an individualized multimodal treatment approach including psychoeducation, pharmacological interventions, and non-pharmacological interventions. Available medications include stimulants (methylphenidate, amphetamines) and non-stimulants (atomoxetine, guanfacine, clonidine). While available pharmacological treatment options for ADHD show relatively large effect sizes (in short-term trials) and overall good tolerability, there is still a need for improvement of current pharmacotherapeutic strategies and for the development of novel medications. This review summarizes available pharmacological treatment options for ADHD in children and adolescents, identifies current issues in research and evidence gaps, and provides an overview of ongoing efforts to develop new medications for the treatment of ADHD in children and adolescents by means of a systematic cross-sectional analysis of the clinical trials registry www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Konstantin Mechler
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Alexander Häge
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
165
|
Borgarelli C, Klingl YE, Escamilla-Ayala A, Munck S, Van Den Bosch L, De Borggraeve WM, Ismalaj E. Lighting Up the Plasma Membrane: Development and Applications of Fluorescent Ligands for Transmembrane Proteins. Chemistry 2021; 27:8605-8641. [PMID: 33733502 DOI: 10.1002/chem.202100296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Indexed: 12/16/2022]
Abstract
Despite the fact that transmembrane proteins represent the main therapeutic targets for decades, complete and in-depth knowledge about their biochemical and pharmacological profiling is not fully available. In this regard, target-tailored small-molecule fluorescent ligands are a viable approach to fill in the missing pieces of the puzzle. Such tools, coupled with the ability of high-precision optical techniques to image with an unprecedented resolution at a single-molecule level, helped unraveling many of the conundrums related to plasma proteins' life-cycle and druggability. Herein, we review the recent progress made during the last two decades in fluorescent ligand design and potential applications in fluorescence microscopy of voltage-gated ion channels, ligand-gated ion channels and G-coupled protein receptors.
Collapse
Affiliation(s)
- Carlotta Borgarelli
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven Campus Arenberg Celestijnenlaan 200F -, box 2404, 3001, Leuven, Belgium
| | - Yvonne E Klingl
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Neurobiology, VIB, Center for Brain &, Disease Research, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Abril Escamilla-Ayala
- Center for Brain & Disease Research, & VIB BioImaging Core, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Department of Neurosciences, Leuven Brain Institute, KU Leuven, Campus Gasthuisberg O&N5 - box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Sebastian Munck
- Center for Brain & Disease Research, & VIB BioImaging Core, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Department of Neurosciences, Leuven Brain Institute, KU Leuven, Campus Gasthuisberg O&N5 - box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Neurobiology, VIB, Center for Brain &, Disease Research, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Wim M De Borggraeve
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven Campus Arenberg Celestijnenlaan 200F -, box 2404, 3001, Leuven, Belgium
| | - Ermal Ismalaj
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven Campus Arenberg Celestijnenlaan 200F -, box 2404, 3001, Leuven, Belgium
| |
Collapse
|
166
|
Bueno TC, Collaço RDC, Cardoso BA, Bredariol RF, Escobar ML, Cajado IB, Gracia M, Antunes E, Zambelli VO, Picolo G, Cury Y, Morandini AC, Marques AC, Sciani JM, Rocha T. Neurotoxicity of Olindias sambaquiensis and Chiropsalmus quadrumanus extracts in sympathetic nervous system. Toxicon 2021; 199:127-138. [PMID: 34139257 DOI: 10.1016/j.toxicon.2021.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 10/21/2022]
Abstract
Cnidarians are equipped with nematocysts, which are specialized organelles used to inoculate venom during prey capturing and defense. Their venoms are rich in toxins and a potential source of bioactive compounds, however, poorly explored so far. In this work, the activity of the methanolic extracts from the hydromedusa Olindias sambaquiensis and the cubozoan jellyfish Chiropsalmus quadrumanus were studied in sympathetic neurotransmission. For that, bisected rat vas deferens - a classic model of sympathetic neurotransmission - were incubated with the extracts for further myographic and histopathological analysis. The O. sambaquiensis extract, at 0.1 μg/mL, facilitated the neurogenic contractions of the noradrenergic-rich epididymal portion, while reducing the noradrenaline (NA) potency, which suggests an interaction with postsynaptic α1-adrenoceptors. On the other hand, a higher concentration (1 μg/mL) leads to time- and frequency-dependent blockade of nerve-evoked contractions without significantly changing the response to exogenous NA. In turn, the C. quadrumanus extract at 0.1 μg/mL induced blockade of nerve-evoked noradrenergic contractions while reducing the potency to exogenous NA. Both extracts did not affect the purinergic neurotransmission or induce muscle damages. Our results demonstrate that O. sambaquiensis and C. quadrumanus extracts significantly interfere with the noradrenergic neurotransmission without altering purinergic response or smooth muscle structure on rat vas deferens. Such results bring to light the pharmacological potential of O. sambaquiensis and C. quadrumanus molecules for therapeutics focusing on noradrenergic neurotransmission.
Collapse
Affiliation(s)
- Thais Cavenatti Bueno
- Multidisciplinary Research Laboratory, São Francisco University (USF), Avenida São Francisco de Assis, 218, Jardim São José, 12916-900, Bragança Paulista, SP, Brazil.
| | - Rita de Cássia Collaço
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz, 13083-887, Campinas, SP, Brazil.
| | - Bianca Aparecida Cardoso
- Multidisciplinary Research Laboratory, São Francisco University (USF), Avenida São Francisco de Assis, 218, Jardim São José, 12916-900, Bragança Paulista, SP, Brazil.
| | - Rafael Fumachi Bredariol
- Multidisciplinary Research Laboratory, São Francisco University (USF), Avenida São Francisco de Assis, 218, Jardim São José, 12916-900, Bragança Paulista, SP, Brazil.
| | - Marília Leal Escobar
- Multidisciplinary Research Laboratory, São Francisco University (USF), Avenida São Francisco de Assis, 218, Jardim São José, 12916-900, Bragança Paulista, SP, Brazil.
| | - Isabela Bubenik Cajado
- Multidisciplinary Research Laboratory, São Francisco University (USF), Avenida São Francisco de Assis, 218, Jardim São José, 12916-900, Bragança Paulista, SP, Brazil.
| | - Marta Gracia
- Multidisciplinary Research Laboratory, São Francisco University (USF), Avenida São Francisco de Assis, 218, Jardim São José, 12916-900, Bragança Paulista, SP, Brazil.
| | - Edson Antunes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz, 13083-887, Campinas, SP, Brazil.
| | - Vanessa O Zambelli
- Laboratory of Pain and Signaling, Butantan Institute, Avenida Vital Brasil, 1500, 05503-900, São Paulo, SP, Brazil.
| | - Gisele Picolo
- Laboratory of Pain and Signaling, Butantan Institute, Avenida Vital Brasil, 1500, 05503-900, São Paulo, SP, Brazil.
| | - Yara Cury
- Laboratory of Pain and Signaling, Butantan Institute, Avenida Vital Brasil, 1500, 05503-900, São Paulo, SP, Brazil.
| | - André C Morandini
- Department of Zoology, Institute of Biosciences, University of São Paulo, São Paulo, 05508-090, Brazil; Marine Biology Center, University of São Paulo, São Sebastião, 11612-109, Brazil.
| | - Antonio C Marques
- Department of Zoology, Institute of Biosciences, University of São Paulo, São Paulo, 05508-090, Brazil.
| | - Juliana Mozer Sciani
- Multidisciplinary Research Laboratory, São Francisco University (USF), Avenida São Francisco de Assis, 218, Jardim São José, 12916-900, Bragança Paulista, SP, Brazil.
| | - Thalita Rocha
- Multidisciplinary Research Laboratory, São Francisco University (USF), Avenida São Francisco de Assis, 218, Jardim São José, 12916-900, Bragança Paulista, SP, Brazil.
| |
Collapse
|
167
|
Bozorgi H, Zamani M, Motaghi E, Eslami M. Dexmedetomidine as an Analgesic Agent with Neuroprotective Properties: Experimental and Clinical Aspects. J Pain Palliat Care Pharmacother 2021; 35:215-225. [PMID: 34100671 DOI: 10.1080/15360288.2021.1914280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Dexmedetomidine (Dexdor or Precedex®) is considered as a sedative agent which is widely used as an adjuvant in general anesthesia and critical care practice. There is extensive evidence indicating its neuroprotective properties especially in various ischemic and hemorrhagic brain injury models of animals. Clinical trials have shown that dexmedetomidine (DEX) can improve the outcome of intensive care unit (ICU) patients. Also, DEX is appropriate as a non-opioid analgesic therapy whenever minimizing opioid-related side effects is necessary. The present article reviews the recent advances in the use of DEX as a neuroprotective agent in both animal and human studies including newest findings about the mechanism of the drug as well as analgesic efficacy of this drug at all perioperative stages. In spite of the beneficial effects of the drug on the nervous system, there are potential adverse effects, such as hypotension and bradycardia, which can be treated pharmacologically and must be taken into consideration by clinicians.
Collapse
Affiliation(s)
- Hooman Bozorgi
- Hooman Bozorgi is with the Department of Pharmacology, Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran. Melika Zamani is with the Department of Pharmacology, School of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran/Panzdah-e-Khordad Hospital, Mahdishahr, Iran. Ehsan Motaghi is with the Department of Physiology and Pharmacology, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran. Majid Eslami is with Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Melika Zamani
- Hooman Bozorgi is with the Department of Pharmacology, Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran. Melika Zamani is with the Department of Pharmacology, School of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran/Panzdah-e-Khordad Hospital, Mahdishahr, Iran. Ehsan Motaghi is with the Department of Physiology and Pharmacology, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran. Majid Eslami is with Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Ehsan Motaghi
- Hooman Bozorgi is with the Department of Pharmacology, Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran. Melika Zamani is with the Department of Pharmacology, School of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran/Panzdah-e-Khordad Hospital, Mahdishahr, Iran. Ehsan Motaghi is with the Department of Physiology and Pharmacology, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran. Majid Eslami is with Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Eslami
- Hooman Bozorgi is with the Department of Pharmacology, Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran. Melika Zamani is with the Department of Pharmacology, School of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran/Panzdah-e-Khordad Hospital, Mahdishahr, Iran. Ehsan Motaghi is with the Department of Physiology and Pharmacology, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran. Majid Eslami is with Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
168
|
Shanahan E, Ryan S, Leahy A, Sheehy T, Costelloe A, Roy A, Galvin R, Peters C, Lyons D, O'Connor M. Delirium and abnormal autonomic nervous system response to head-up tilt testing. Exp Gerontol 2021; 152:111430. [PMID: 34102273 DOI: 10.1016/j.exger.2021.111430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/14/2021] [Accepted: 05/27/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Delirium is a common condition with poorly understood pathophysiology. Various theories have been proposed including that delirious patients have reduced cerebral blood flow. We hypothesised that patients with delirium could have abnormal autonomic nervous system function, as assessed by tilt table testing, which would explain the alteration in blood flow. METHODS A prospective cohort study of medical inpatients aged 65 years and older was undertaken. Delirium was assessed using DRS-R98 and DSM-IV criteria. Beat-to-beat blood pressure (BP) was recorded during tilt testing. Differences in BP changes between the two groups (those with delirium and those without) were explored. The association between severity of delirium and magnitude of BP changes was also examined. RESULTS 64 participants were recruited during hospitalisation. 29 completed follow-up Head-Up Tilt testing. The mean age of participants was 80.8 years (SD 6.2 years). The control group (n = 12) had a median decrease in systolic BP of 17.5 mmHg (IQR 20.75). The delirium group (n = 17) had a median decrease in systolic BP of 1 mmHg (IQR 38.5), p = 0.04. As delirium severity scores increased, systolic BP change during tilting also increased (rs = 0.42, p = 0.03). CONCLUSION Participants in the delirium group showed different BP responses to tilt test which may represent abnormal sympathetic response. This would be consistent with other features of delirium such as treatment response to centrally acting alpha-2 blockers. Equity of access to research for older, frail and delirious cohorts is essential but feasibility and acceptability needs to be optimised and factored into study design.
Collapse
Affiliation(s)
- Elaine Shanahan
- Department of Ageing and Therapeutics, University Hospital Limerick, Ireland.
| | - Sheila Ryan
- Department of Ageing and Therapeutics, University Hospital Limerick, Ireland
| | - Aoife Leahy
- Department of Ageing and Therapeutics, University Hospital Limerick, Ireland; School of Allied Health, Faculty of Education and Health Sciences, Ageing Research Institute, Health Research Institute, University of Limerick, Ireland
| | - Tina Sheehy
- Department of Ageing and Therapeutics, University Hospital Limerick, Ireland
| | - Aine Costelloe
- Department of Ageing and Therapeutics, University Hospital Limerick, Ireland
| | - Amrita Roy
- Mater Misericordiae University Hospital, Dublin, Ireland
| | - Rose Galvin
- School of Allied Health, Faculty of Education and Health Sciences, Ageing Research Institute, Health Research Institute, University of Limerick, Ireland
| | - Catherine Peters
- Department of Ageing and Therapeutics, University Hospital Limerick, Ireland
| | - Declan Lyons
- Department of Ageing and Therapeutics, University Hospital Limerick, Ireland
| | - Margaret O'Connor
- Department of Ageing and Therapeutics, University Hospital Limerick, Ireland
| |
Collapse
|
169
|
Liu Y, Ai D, Wang X. Efficacy of perioperative intravenous dexmedetomidine administration for the prevention of postoperative sore throat: a meta-analysis. J Int Med Res 2021; 49:3000605211017686. [PMID: 34044638 PMCID: PMC8165843 DOI: 10.1177/03000605211017686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective Postoperative sore throat (POST) is an undesirable intubation-related
complication after surgery. Several studies have investigated the efficacy
of perioperative intravenous dexmedetomidine administration for the
prevention of POST, but the results have been inconsistent. We aimed to
summarize all existing evidence and draw a more precise conclusion to guide
future clinical work. Methods PubMed, Cochrane Library, EMBASE and China National Knowledge Infrastructure
databases were comprehensively searched for all randomized controlled trials
published before 1 February 2021 that investigated the efficacy of
dexmedetomidine for the prevention of POST. Results Nine studies involving 400 patients were included in our meta-analysis.
Compared with the control groups (i.e., saline and anesthetic drugs),
perioperative intravenous use of dexmedetomidine significantly reduced the
incidence of POST [risk ratio (RR): 0.56; 95% confidence interval (CI):
0.40–0.77; I2 = 0%) and coughing on the tube
during extubation (RR: 0.58; 95% CI: 0.41–0.82;
I2 = 0%). Additionally, patients in the
dexmedetomidine group were more likely to develop bradycardia (RR: 2.46; 95%
CI: 1.28–4.71; I2 = 0%) and hypotension (RR:
3.26; 95% CI: 1.14–9.33; I2 = 0%) during the
administration of dexmedetomidine than those in the control group. Conclusion Perioperative intravenous administration of dexmedetomidine has a positive
effect on the prevention of POST.
Collapse
Affiliation(s)
- Yuanhui Liu
- Department of Anesthesiology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Dongmei Ai
- Department of Anesthesiology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaobin Wang
- Department of Anesthesiology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
170
|
Mwobobia RM, Kanui TI, Abelson KSP. The effects of clonidine and yohimbine in the tail flick and hot plate tests in the naked mole rat (Heterocephalus glaber). BMC Res Notes 2021; 14:191. [PMID: 34001271 PMCID: PMC8130107 DOI: 10.1186/s13104-021-05607-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/08/2021] [Indexed: 11/10/2022] Open
Abstract
Objective The naked mole rat (NMR) (Heterocephalus glaber) is increasingly considered an important biomedical research model for various conditions like hypoxic brain injury, cancer and nociception. This study was designed to investigate the effects of clonidine and yohimbine, an alpha-2 (α2) adrenoceptor agonist and antagonist respectively in the tail flick and hot plate tests. Results A significant difference in tail flick latency was noted between saline control and 30 µg/kg clonidine, which was reduced after administration of 30 µg/kg yohimbine. A significant difference in hot plate latency was also noted between saline control and 30 µg/kg clodinine during the periods 30, 45, 60, 75 and 90 min after administration, and between saline control and 10 µg/kg clonidine during 30 min after administration. The hot plate latency by 30 µg/kg clonidine was also reduced by 30 µg/kg yohimbine during 30 min after administration. Since the tail-flick and hot plate tests mediate the effects at spinal and supraspinal levels respectively, the present study indicates the presence and involvement of noradrenergic receptors in thermal antinociception at spinal and supraspinal levels of the NMR, similar to what has been found in other mammals.
Collapse
Affiliation(s)
- R M Mwobobia
- School of Agriculture and Veterinary Sciences, South Eastern Kenya University, P O Box 170-90200, Kitui, Kenya. .,Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamvej 3B, 2200, Copenhagen, Denmark.
| | - T I Kanui
- School of Agriculture and Veterinary Sciences, South Eastern Kenya University, P O Box 170-90200, Kitui, Kenya
| | - K S P Abelson
- Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamvej 3B, 2200, Copenhagen, Denmark
| |
Collapse
|
171
|
Bi X, Wei J, Zhang X. Effects of dexmedetomidine on neurocognitive disturbance after elective non-cardiac surgery in senile patients: a systematic review and meta-analysis. J Int Med Res 2021; 49:3000605211014294. [PMID: 33983077 PMCID: PMC8371033 DOI: 10.1177/03000605211014294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Senile patients often experience neurocognitive disturbance after non-cardiac surgery. Several clinical trials have investigated if the perioperative intravenous use of dexmedetomidine has a positive effect on the prevention of neurocognitive dysfunction, but the results have been inconsistent. We performed a meta-analysis to investigate the effects of dexmedetomidine on neurocognitive disturbance after elective non-cardiac surgery in senile patients. METHODS The PubMed, Cochrane Library, EMBASE and China National Knowledge Infrastructure databases were comprehensively searched for all randomized controlled trials published before 1 February 2020 that investigated the efficacy of dexmedetomidine in the prevention of postoperative delirium (POD) or postoperative cognitive dysfunction (POCD). RESULTS Sixteen studies involving 4376 patients were included in this meta-analysis. Compared with the control (i.e., saline), the perioperative intravenous use of dexmedetomidine significantly reduced the incidence of POD and POCD. However, patients in the dexmedetomidine group were more likely to develop bradycardia and hypotension during the administration of dexmedetomidine than patients in the control group. There were no differences between the two groups in the incidence of nausea and vomiting or mortality rate. CONCLUSION Dexmedetomidine has a positive effect on the prevention of neurocognitive disturbance in senile patients after elective non-cardiac surgery.
Collapse
Affiliation(s)
- Xiaobo Bi
- Department of Anesthesiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jingxia Wei
- Department of Pharmacology, the Sixth People's Hospital of Chengdu, Chengdu, Sichuan China
| | - Xia Zhang
- Department of Neonatology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
172
|
Jarrar YB, Jarrar Q, Abaalkhail SJ, Moh'd Kalloush H, Naser W, Zihlif M, Al Shhab M, El Madani A, Jamous Y, Lee SJ. Molecular toxicological alterations in the mouse hearts induced by sub-chronic thiazolidinedione drugs administration. Fundam Clin Pharmacol 2021; 36:143-149. [PMID: 33969534 DOI: 10.1111/fcp.12694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022]
Abstract
Thiazolidinediones are well-known anti-diabetic drugs. However, they are not widely used due to their cardiotoxic effects. Therefore, in this study, we aimed to determine the molecular toxicological alterations induced in the mouse hearts after thiazolidinedione administration. Balb/c mice received doses clinically equivalent to those given to humans of the most commonly used thiazolidinediones, pioglitazone, and rosiglitazone for 30 days. After that, RNA samples were isolated from the hearts. The mRNA expression of cytochrome (cyp) p450 genes that synthesize the cardiotoxic 20-hydroxyeicosatetraenoic acid (20-HETE) in addition to 92 cardiotoxicity biomarker genes were analyzed using quantitative polymerase chain reaction array technique. The analysis demonstrated that thiazolidinediones caused a significant upregulation (p < 0.5) of the mRNA expression of cyp1a1, cyp4a12, itpr1, ccl7, ccr1, and b2 m genes. In addition, thiazolidinediones caused a significant (p < 0.05) downregulation of the mRNA expression of adra2a, bsn, col15a1, fosl1, Il6, bpifa1, plau, and reg3b genes. The most affected gene was itpr1 gene, which was upregulated by pioglitazone and rosiglitazone by sevenfold and 3.5-fold, respectively. In addition, pioglitazone caused significant upregulation of (p < 0.05) hamp, ppbp, psma2, sik1, timp1, and ucp1 genes, which were not affected significantly (p > 0.05) by rosiglitazone administration. In conclusion, this study showed that thiazolidinediones induce toxicological molecular alterations in the mouse hearts, such as the induction of cyp450s that synthesize 20-HETE, chemokine activation, inflammatory responses, blood clotting, and oxidative stress. These findings may help us understand the mechanism of cardiotoxicity involved in thiazolidinedione administration.
Collapse
Affiliation(s)
| | - Qais Jarrar
- Department of Pharmaceutical Science, Al-Isra'a University, Amman, Jordan
| | - Sara J Abaalkhail
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | | | - Wisam Naser
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Malek Zihlif
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Mohammad Al Shhab
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Abdulla El Madani
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Yahya Jamous
- Nanotechnology Centre, King AbdulAziz City of Science and Technology, Riyadh, Saudi Arabia
| | - Su-Jun Lee
- Department of Pharmacology, College of Medicine, Inje University, Busan, South Korea
| |
Collapse
|
173
|
Obeng S, Hiranita T, León F, McMahon LR, McCurdy CR. Novel Approaches, Drug Candidates, and Targets in Pain Drug Discovery. J Med Chem 2021; 64:6523-6548. [PMID: 33956427 DOI: 10.1021/acs.jmedchem.1c00028] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Because of the problems associated with opioids, drug discovery efforts have been employed to develop opioids with reduced side effects using approaches such as biased opioid agonism, multifunctional opioids, and allosteric modulation of opioid receptors. Receptor targets such as adrenergic, cannabinoid, P2X3 and P2X7, NMDA, serotonin, and sigma, as well as ion channels like the voltage-gated sodium channels Nav1.7 and Nav1.8 have been targeted to develop novel analgesics. Several enzymes, such as soluble epoxide hydrolase, sepiapterin reductase, and MAGL/FAAH, have also been targeted to develop novel analgesics. In this review, old and recent targets involved in pain signaling and compounds acting at these targets are summarized. In addition, strategies employed to reduce side effects, increase potency, and efficacy of opioids are also elaborated. This review should aid in propelling drug discovery efforts to discover novel analgesics.
Collapse
Affiliation(s)
- Samuel Obeng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States.,Department Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Takato Hiranita
- Department Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Francisco León
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia 29208, United States
| | - Lance R McMahon
- Department Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States.,Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
174
|
Egbuta C, Mason KP. Current State of Analgesia and Sedation in the Pediatric Intensive Care Unit. J Clin Med 2021; 10:1847. [PMID: 33922824 PMCID: PMC8122992 DOI: 10.3390/jcm10091847] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/15/2022] Open
Abstract
Critically ill pediatric patients often require complex medical procedures as well as invasive testing and monitoring which tend to be painful and anxiety-provoking, necessitating the provision of analgesia and sedation to reduce stress response. Achieving the optimal combination of adequate analgesia and appropriate sedation can be quite challenging in a patient population with a wide spectrum of ages, sizes, and developmental stages. The added complexities of critical illness in the pediatric population such as evolving pathophysiology, impaired organ function, as well as altered pharmacodynamics and pharmacokinetics must be considered. Undersedation leaves patients at risk of physical and psychological stress which may have significant long term consequences. Oversedation, on the other hand, leaves the patient at risk of needing prolonged respiratory, specifically mechanical ventilator, support, prolonged ICU stay and hospital admission, and higher risk of untoward effects of analgosedative agents. Both undersedation and oversedation put critically ill pediatric patients at high risk of developing PICU-acquired complications (PACs) like delirium, withdrawal syndrome, neuromuscular atrophy and weakness, post-traumatic stress disorder, and poor rehabilitation. Optimal analgesia and sedation is dependent on continuous patient assessment with appropriately validated tools that help guide the titration of analgosedative agents to effect. Bundled interventions that emphasize minimizing benzodiazepines, screening for delirium frequently, avoiding physical and chemical restraints thereby allowing for greater mobility, and promoting adequate and proper sleep will disrupt the PICU culture of immobility and reduce the incidence of PACs.
Collapse
Affiliation(s)
| | - Keira P. Mason
- Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA;
| |
Collapse
|
175
|
Dowdy RAE, Mansour ST, Cottle JH, Mabe HR, Weprin HB, Yarborough LE, Ness GM, Jacobs TM, Cornelius BW. Cardiac Arrest Upon Induction of General Anesthesia. Anesth Prog 2021; 68:38-44. [PMID: 33827129 DOI: 10.2344/anpr-67-03-08] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 06/02/2020] [Indexed: 11/11/2022] Open
Abstract
There are numerous causes of cardiac arrest in the perioperative period, including hypoxia, hypovolemia, and vagal response to medications or procedures during routine anesthetics. Initiation of adequate cardiopulmonary resuscitation, administration of epinephrine, and application of a defibrillator, with shocking when applicable, are all essential steps in achieving return of spontaneous circulation. Knowledge and utilization of monitoring equipment can alert the provider to problems leading to cardiac arrest as well as ensure proper resuscitative efforts during the event. Polypharmacy is quite common with many of today's special needs patients. It is important to understand the medications they are taking as well as the potential interactions that may occur with drugs given during sedation and general anesthesia. The following is a case report of cardiac arrest including asystole and pulseless electrical activity in a 27-year-old man with autism and behavioral problems who presented for restorative dentistry under general anesthesia in the ambulatory surgery setting.
Collapse
Affiliation(s)
- Regina A E Dowdy
- Division of Oral and Maxillofacial Surgery and Dental Anesthesiology, The Ohio State University, Columbus, Ohio
| | | | - James H Cottle
- College of Dentistry, The Ohio State University, Columbus, Ohio
| | - Hannah R Mabe
- College of Dentistry, The Ohio State University, Columbus, Ohio
| | - Harry B Weprin
- College of Dentistry, The Ohio State University, Columbus, Ohio
| | | | - Gregory M Ness
- Division of Oral and Maxillofacial Surgery and Dental Anesthesiology, The Ohio State University, Columbus, Ohio
| | - Todd M Jacobs
- Division of Oral and Maxillofacial Surgery and Dental Anesthesiology, The Ohio State University, Columbus, Ohio
| | - Bryant W Cornelius
- Division of Oral and Maxillofacial Surgery and Dental Anesthesiology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
176
|
Zhang BP, Wu L, Wu XW, Wang F, Zhao X. Dexmedetomidine protects against degeneration of dopaminergic neurons and improves motor activity in Parkinson's disease mice model. Saudi J Biol Sci 2021; 28:3198-3203. [PMID: 34121856 PMCID: PMC8176059 DOI: 10.1016/j.sjbs.2021.04.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/04/2021] [Accepted: 04/05/2021] [Indexed: 11/18/2022] Open
Abstract
Parkinson’s disease (PD) is the result of dopaminergic (DA) neuronal death in the substantianigra pars compacta (SNc). Current treatments for PD such as L-dopa are limited in effectiveness and fail to address the cause. Targeted anti-inflammatory therapies, particularly directed at nuclear factor kappa B (NF‐κB) activity in alleviating degeneration of DA-neurons is of evolving interest. In the present study, we hypothesised that dexmedetomidine (DEX), an alpha-2 receptor adrenergic agonist, suppress the inflammatory responses associated with PD and restores dopaminergic levels by alleviating substantia nigral degeneration. Male mice (C57Bl/10, 8–11 months old and of 34–40 g of weight) were divided into: the control, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and MPTP + dexmedetomidine (MPTP + DEX) (n = 26 each group). Dex restored dopamine levels in SNpc of MPTP-induced PD mice model. Results of immunohisto staining revealed that Dex treatment post-MPTP induction restored TH-positive cells, with only 12.37% increase (##p < 0.01 vs MPTP) on the third day and a steep 55% increase (###p < 0.001 vs MPTP) following the seventh day of Dex treatment. Moreover, the expressions of proinflammatory markers regulated by NF-κB were diminished in Dex + MPTP group. In addition, cylinder test revealed that Dex treatment improved asymmetric limb usage pattern in MPTP induced mice over the course of 7 days. Hence, in this study, we provided insight on the effect of Dex in the inhibition of NF-κB1 regulated proinflammatory mediators to improve dopamine levels and reduce SNpc dopaminergic neuronal degeneration.
Collapse
Affiliation(s)
- Bao-Ping Zhang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054 Shaanxi, China
| | - Li Wu
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054 Shaanxi, China
| | - Xian-Wei Wu
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054 Shaanxi, China
| | - Fang Wang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054 Shaanxi, China
| | - Xin Zhao
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054 Shaanxi, China
| |
Collapse
|
177
|
Colombe AS, Pidoux G. Cardiac cAMP-PKA Signaling Compartmentalization in Myocardial Infarction. Cells 2021; 10:cells10040922. [PMID: 33923648 PMCID: PMC8073060 DOI: 10.3390/cells10040922] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/02/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
Under physiological conditions, cAMP signaling plays a key role in the regulation of cardiac function. Activation of this intracellular signaling pathway mirrors cardiomyocyte adaptation to various extracellular stimuli. Extracellular ligand binding to seven-transmembrane receptors (also known as GPCRs) with G proteins and adenylyl cyclases (ACs) modulate the intracellular cAMP content. Subsequently, this second messenger triggers activation of specific intracellular downstream effectors that ensure a proper cellular response. Therefore, it is essential for the cell to keep the cAMP signaling highly regulated in space and time. The temporal regulation depends on the activity of ACs and phosphodiesterases. By scaffolding key components of the cAMP signaling machinery, A-kinase anchoring proteins (AKAPs) coordinate both the spatial and temporal regulation. Myocardial infarction is one of the major causes of death in industrialized countries and is characterized by a prolonged cardiac ischemia. This leads to irreversible cardiomyocyte death and impairs cardiac function. Regardless of its causes, a chronic activation of cardiac cAMP signaling is established to compensate this loss. While this adaptation is primarily beneficial for contractile function, it turns out, in the long run, to be deleterious. This review compiles current knowledge about cardiac cAMP compartmentalization under physiological conditions and post-myocardial infarction when it appears to be profoundly impaired.
Collapse
|
178
|
Gezalian MM, Mangiacotti L, Rajput P, Sparrow N, Schlick K, Lahiri S. Cerebrovascular and neurological perspectives on adrenoceptor and calcium channel modulating pharmacotherapies. J Cereb Blood Flow Metab 2021; 41:693-706. [PMID: 33210576 PMCID: PMC7983505 DOI: 10.1177/0271678x20972869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 02/02/2023]
Abstract
Adrenoceptor and calcium channel modulating medications are widely used in clinical practice for acute neurological and systemic conditions. It is generally assumed that the cerebrovascular effects of these drugs mirror that of their systemic effects - and this is reflected in how these medications are currently used in clinical practice. However, recent research suggests that there are distinct cerebrovascular-specific effects of these medications that are related to the unique characteristics of the cerebrovascular anatomy including the regional heterogeneity in density and distribution of adrenoceptor subtypes and calcium channels along the cerebrovasculature. In this review, we critically evaluate existing basic science and clinical research to discuss known and putative interactions between adrenoceptor and calcium channel modulating pharmacotherapies, the neurovascular unit, and cerebrovascular anatomy. In doing so, we provide a rationale for selecting vasoactive medications based on lesion location and lay a foundation for future investigations that will define neuroprotective paradigms of adrenoceptor and calcium channel modulating therapies to improve neurological outcomes in acute neurological and systemic disorders.
Collapse
Affiliation(s)
- Michael M Gezalian
- Departments of Neurology and Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Luigi Mangiacotti
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Padmesh Rajput
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nicklaus Sparrow
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Konrad Schlick
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shouri Lahiri
- Departments of Neurology, Neurosurgery, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
179
|
McAdams RM, Berube MW. Emerging therapies and management for neonatal encephalopathy-controversies and current approaches. J Perinatol 2021; 41:661-674. [PMID: 33712717 DOI: 10.1038/s41372-021-01022-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 01/31/2023]
Abstract
Neonatal encephalopathy (NE) continues to have a major impact on newborn survival and neurodevelopmental outcomes worldwide. In high-income settings, therapeutic hypothermia is the only established standard treatment for neonates with moderate-to-severe NE, with compelling evidence that cooling reduces mortality and major neurodevelopmental impairment in survivors. Despite therapeutic hypothermia, a significant proportion of cooled infants continue to suffer long-term disability from brain injury. Innovative therapies offer the possibility of further improving neurodevelopmental outcomes by working synergistically with therapeutic hypothermia to decrease hypoxia-ischemia-induced excitotoxicity, prevent progression to secondary energy failure, and in some cases, promote neuroregeneration in the developing neonatal brain. This review discusses emerging NE therapies currently under investigation, offers insight into controversies surrounding various approaches to clinical care during therapeutic hypothermia, and identifies ongoing knowledge deficits that hinder attainment of optimal outcomes for neonates with NE.
Collapse
Affiliation(s)
- Ryan M McAdams
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Megan W Berube
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
180
|
The addition of clonidine to ropivacaine in rectus sheath nerve blocks for pediatric patients undergoing laparoscopic appendectomy: A double blinded randomized prospective study. J Clin Anesth 2021; 71:110254. [PMID: 33752119 DOI: 10.1016/j.jclinane.2021.110254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 11/23/2022]
Abstract
STUDY OBJECTIVE The primary goal of this study was to determine if the addition of clonidine to ropivacaine prolonged periumbilical numbness compared to ropivacaine alone in pediatric patients receiving ultrasound guided rectus sheath nerve blocks for laparoscopic appendectomy. The secondary goals were to evaluate differences in perioperative pain scores, analgesic consumption, sedation, anxiolysis, and hemodynamic effects from clonidine. DESIGN This was a single center, randomized, double-blinded prospective study. SETTING This study was conducted within the pediatric operating rooms at the Children's Hospital of Pittsburgh, a large university-based academic medical center. PATIENTS Fifty pediatric patients (ages 10-17 years old) without pre-existing cognitive impairment, developmental delay or chronic pain undergoing laparoscopic appendectomy during weekday hours were enrolled and randomized to control versus intervention groups. INTERVENTION Ultrasound guided rectus sheath nerve block injections were performed at the beginning of surgery with either ropivacaine 0.5% plus normal saline or ropivacaine 0.5% plus clonidine (2 mcg/kg, maximum of 100 mcg). MEASUREMENTS The duration of periumbilical numbness, Numeric Pain Rating Scale scores, University of Michigan Sedation Scale, State-Trait Anxiety Inventory for Children, analgesic consumption, heart rate, blood pressure, and mean arterial pressures, were recorded for each patient at several time points in the perioperative setting. MAIN RESULTS There were no significant differences in demographic characteristics between groups. The median duration of periumbilical numbness did not significantly differ between the ropivacaine only and the ropivacaine plus clonidine groups 540.0 minutes [360.0 -1015.0] (median [interquartile range (IQR)]) versus 823.5 minutes [509.5- 1080.0], p = 0.451. There were no significant differences in perioperative analgesic consumption, pain and anxiety scores, PACU sedation, or hemodynamic instability. CONCLUSIONS The addition of clonidine did not significantly prolong rectus sheath nerve block duration and was well tolerated in pediatric patients. Perioperative analgesia, hemodynamics, anxiety, and PACU sedation did not differ between groups. TRIAL REGISTRATION Clinical Trials NCT02439281.
Collapse
|
181
|
Killoran KE, Walsh CA, Asher JL, Tarleton MB, Wilson SR. Rapid Recovery and Short Duration Anesthesia after Low Dose Ketamine and High Dose Dexmedetomidine in Rhesus Macaques ( Macaca mulatta). JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE 2021; 60:337-340. [PMID: 33731246 DOI: 10.30802/aalas-jaalas-20-000112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Anesthesia in rhesus macaques is required for many procedures. Although ketamine is the backbone of most anesthetic protocols, tolerance to the drug can develop, resulting in the need for higher doses to provide sufficient restraint. Combination with other drugs, such as α-agonists, can be ketamine-sparing, providing for sufficient restraint at lower ketamine doses. In addition, because α-agonists are reversible, recovery from anesthesia has the potential to be much shorter. We hypothesized that use of a low dose of ketamine with a high dose of dexmedetomidine, an α2 receptor selective agonist, in male and female rhesus macaques less than 15 y of age would provide adequate anesthesia for short procedures and that recovery would be faster than in macaques given a higher dose of ketamine (10 mg/kg) alone. We found that the combination, in conjunction with atipamezole for reversal, provided smooth induction of anesthesia and significantly shorter recovery time than did ketamine alone, with no significant effects of sex. The combination of low dose ketamine and high dose dexmedetomidine also provided a 30-min window of anesthesia with analgesia sufficient for mild to moderately painful procedures.
Collapse
Affiliation(s)
- Kristin E Killoran
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Courtney A Walsh
- Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| | - Jennifer L Asher
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Molly B Tarleton
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Steven R Wilson
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut;,
| |
Collapse
|
182
|
Fortune S, Frawley J. Optimizing Pain Control and Minimizing Opioid Use in Trauma Patients. AACN Adv Crit Care 2021; 32:89-104. [PMID: 33725102 DOI: 10.4037/aacnacc2021519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Adverse effects of opioids and the ongoing crisis of opioid abuse have prompted providers to reduce prescribing opioids and increase use of multiple nonpharmacologic therapies, nonopioid analgesics, and co-analgesics for pain management in trauma patients. Nonopioid agents, including acetaminophen, nonsteroidal anti-inflammatory drugs, gabapentinoids, ketamine, central α2 agonists, and lidocaine, can be used as adjuncts or alternatives to opioids in the trauma population. Complementary therapies such as acupuncture, virtual reality, and mirror therapy are modalities that also may be helpful in reducing pain. Performing pain assessments is fundamental to identify pain and evaluate treatment effectiveness in the critically ill trauma patient. The efficacy, safety, and availability of opioid-sparing therapies and multimodal pain regimens are reviewed.
Collapse
Affiliation(s)
- Shanna Fortune
- Shanna Fortune is Advanced Practice Registered Nurse, Trauma Acute Pain Management Service, R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, Maryland
| | - Jennifer Frawley
- Jennifer Frawley is Trauma Critical Care Clinical Pharmacy Specialist, University of Maryland Medical Center, 22 S Greene St, Baltimore, MD 21201
| |
Collapse
|
183
|
Wu Z, Yu J, Lin Q, Li H, Zhang T, Tan H, Lin W, Cao L. Effects of an Intraoperative Intravenous Bolus Dose of Dexmedetomidine on Remifentanil-Induced Postinfusion Hyperalgesia in Patients Undergoing Thyroidectomy: A Double-Blind Randomized Controlled Trial. Anesth Analg 2021; 132:320-328. [PMID: 32639389 DOI: 10.1213/ane.0000000000005003] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Consecutive exposure to high-dose remifentanil during anesthesia may induce remifentanil-induced postinfusion hyperalgesia (RPH). Dexmedetomidine, a highly selective α2-adrenergic receptor agonist, may have synergistic effects with opioids and aid in perioperative pain management. In this study, we hypothesized that an intraoperative bolus dose of intravenous dexmedetomidine could alleviate RPH in patients undergoing thyroidectomy under general anesthesia. METHODS Ninety patients undergoing thyroidectomy were randomly assigned to 1 of 3 groups: placebo, normal saline (group P); low-dose dexmedetomidine 0.2 μg·kg-1 (group LD); or high-dose dexmedetomidine 0.5 μg·kg-1 (group HD). Remifentanil was infused at a rate of 0.30 μg·kg-1·minute-1. Mechanical pain thresholds were measured using an Electronic von Frey device preoperatively and at 30 minutes, 6 hours, 24 hours, and 48 hours after surgery and were analyzed with 2-way repeated-measures analysis of variance (ANOVA) followed by Bonferroni post hoc comparison. We also recorded postoperative pain scores, the incidence of receiving rescue analgesics, and side effects up to 48 hours after surgery. RESULTS The mechanical pain thresholds around the skin incision were significantly higher in group LD compared to group P 30 minutes and 6 hours after surgery (mean ± standard deviation: [65.0 ± 25.2] vs [49.6 ± 24.4] g, mean difference [95% confidence interval]: 15.4 [0.3-30.5] g, P = .045 at 30 minutes; [65.9 ± 24.5] vs [49.3 ± 26.1] g, 16.6 [1.1-32.1] g, P = .032 at 6 hours). The pain thresholds around the skin incision were significantly higher in group HD compared to group P 30 minutes and 6 hours after surgery ([67.8 ± 21.7] vs [49.6 ± 24.4] g, 18.2 [3.1-33.3] g, P = .013 at 30 minutes; [68.3 ± 22.5] vs [49.3 ± 26.1] g, 19.0 [3.5-34.5] g, P = .011 at 6 hours). The incidence of hyperalgesia around the skin incision was lower in group HD than in group P 30 minutes and 6 hours after surgery (4 [13%] vs 14 [48%], P = .012 at 30 minutes, 4 [13%] vs 12 [41%], P = .045 at 6 hours), although no significant difference was observed between group LD and group P. Postoperative pain scores, the incidence of rescue analgesic demand, and postoperative side effects were not significantly different between the groups. CONCLUSIONS An intraoperative intravenous bolus dose of dexmedetomidine 0.5 μg·kg-1 alleviates remifentanil-induced hyperalgesia in patients undergoing thyroidectomy without a significant difference in side effects.
Collapse
Affiliation(s)
- Zhijie Wu
- From the Department of Anesthesiology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of Anesthesiology, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong, China
| | - Junjie Yu
- From the Department of Anesthesiology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Qihua Lin
- Department of Anesthesiology, Affiliated Shenzhen Maternal & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Huiting Li
- From the Department of Anesthesiology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Tianhua Zhang
- From the Department of Anesthesiology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Hongying Tan
- From the Department of Anesthesiology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Wenqian Lin
- From the Department of Anesthesiology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Longhui Cao
- From the Department of Anesthesiology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| |
Collapse
|
184
|
Baumgartner K, Mullins M. Pediatric clonidine and guanfacine poisoning: a single-center retrospective review. TOXICOLOGY COMMUNICATIONS 2021. [DOI: 10.1080/24734306.2021.1878322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Affiliation(s)
- Kevin Baumgartner
- Department of Emergency Medicine, Division of Medical Toxicology, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael Mullins
- Department of Emergency Medicine, Division of Medical Toxicology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
185
|
Abstract
Acute kidney injury (AKI) occurs frequently after cardiac surgery and is associated with high morbidity and mortality. Although the number of cardiac surgical procedures is constantly growing worldwide, incidence of cardiac surgery-associated AKI is still around 40% and has a significant impact on global health care costs. Numerous trials attempted to identify strategies to prevent AKI and attenuate its detrimental consequences. Effective options remained elusive. Current evidence supports a multimodal risk-stratification approach with biomarker-guided management of high-risk patients, perioperative administration of dexmedetomidine, and implementation of a care bundle as recommended by the Kidney Disease: Improving Global Outcomes group.
Collapse
|
186
|
El-Tallawy SN, Nalamasu R, Salem GI, LeQuang JAK, Pergolizzi JV, Christo PJ. Management of Musculoskeletal Pain: An Update with Emphasis on Chronic Musculoskeletal Pain. Pain Ther 2021; 10:181-209. [PMID: 33575952 PMCID: PMC8119532 DOI: 10.1007/s40122-021-00235-2] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/09/2021] [Indexed: 12/28/2022] Open
Abstract
Musculoskeletal pain is a challenging condition for both patients and physicians. Many adults have experienced one or more episodes of musculoskeletal pain at some time of their lives, regardless of age, gender, or economic status. It affects approximately 47% of the general population. Of those, about 39–45% have long-lasting problems that require medical consultation. Inadequately managed musculoskeletal pain can adversely affect quality of life and impose significant socioeconomic problems. This manuscript presents a comprehensive review of the management of chronic musculoskeletal pain. It briefly explores the background, classifications, patient assessments, and different tools for management according to the recently available evidence. Multimodal analgesia and multidisciplinary approaches are fundamental elements of effective management of musculoskeletal pain. Both pharmacological, non-pharmacological, as well as interventional pain therapy are important to enhance patient’s recovery, well-being, and improve quality of life. Accordingly, recent guidelines recommend the implementation of preventative strategies and physical tools first to minimize the use of medications. In patients who have had an inadequate response to pharmacotherapy, the proper use of interventional pain therapy and the other alternative techniques are vital for safe and effective management of chronic pain patients.
Collapse
Affiliation(s)
- Salah N El-Tallawy
- Anesthesia and Pain Management Department, King Khalid University Hospital, College of Medicine, King Saud University, Riyadh, Saudi Arabia. .,Faculty of Medicine, Minia University and NCI, Cairo University, Cairo, Egypt.
| | - Rohit Nalamasu
- Department of Physical Medicine and Rehabilitation, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gehan I Salem
- Rheumatology, Rehabilitation and Physical Medicine Department, Assiut University Hospital, Assiut, Egypt.,Rehabilitation Medicine Department, King Khalid University Hospital, King Saud University, Riyadh, Saudi Arabia
| | | | | | - Paul J Christo
- Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
187
|
Lelescu CA, Dumitras DA, Iurian S, Staffieri F, Muresan C. Effects of topical application of tramadol with/without dexmedetomidine and proparacaine on corneal sensitivity in rats. Int Ophthalmol 2021; 41:465-473. [PMID: 33095346 DOI: 10.1007/s10792-020-01596-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/01/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE To evaluate the corneal anesthetic effect following topical application of tramadol alone and in combination with dexmedetomidine, and compare it to proparacaine, in clinically healthy rats. METHODS A randomized, crossover study was performed. Twenty Wistar albino rats (n = 40 eyes) were used. Corneal touch threshold (CTT) measurements (in mm) were obtained using a Cochet-Bonnet aesthesiometer. CTT measurements were obtained at baseline, 1-min following application of the topical anesthetic agent, and repeated at 5-min intervals up to 75 min. The topical protocol involved 3 treatment conditions, separated by a 2-week washout period: proparacaine, tramadol alone, and tramadol in combination with dexmedetomidine. RESULTS CTT values were significantly decreased compared to baseline at each timepoint until completion of the 75-min evaluation in all treated eyes, regardless of the assigned treatment (p < 0.0083). With tramadol, complete corneal anesthesia (CTT = 0) was achieved within 1-5 min in 18 eyes and ranged from 5 to 25 min. Co-administration of dexmedetomidine to tramadol resulted in significantly increased CTT values from 5 to 20 min following topical application, compared to tramadol alone (p < 0.0083), and complete corneal anesthesia was achieved in only 14 out of 20 treated eyes. CONCLUSION Tramadol might be a useful alternative to topical anesthetic agents, providing a dose-related corneal anesthetic effect. Co-administration of dexmedetomidine does not potentiate its anesthetic effect. The underlying mechanism(s) of drug antagonism between tramadol and dexmedetomidine remains to be determined.
Collapse
Affiliation(s)
- Cristina A Lelescu
- Modis Competence Center, Modis Life Sciences Belgium, Mureșului 9, 400000, Cluj-Napoca, Romania
| | - Daria A Dumitras
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3-5, 400372, Cluj-Napoca, Romania
| | - Sonia Iurian
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hațieganu", V. Babeș 41, 400012, Cluj-Napoca, Romania
| | - Francesco Staffieri
- Division of Veterinary Surgery, Department of Emergency and Organ Transplantation, Faculty of Veterinary Medicine, University "Aldo Moro" of Bari, SP Per Casamassima Km. 3, 70010, Valenzano, Bari, Italy
| | - Cosmin Muresan
- Department of Surgery, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3-5, 400372, Cluj-Napoca, Romania.
| |
Collapse
|
188
|
Injection of Lidocaine Alone versus Lidocaine plus Dexmedetomidine in Impacted Third Molar Extraction Surgery, a Double-Blind Randomized Control Trial for Postoperative Pain Evaluation. Pain Res Manag 2021; 2021:6623792. [PMID: 33574974 PMCID: PMC7857915 DOI: 10.1155/2021/6623792] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/24/2020] [Accepted: 01/15/2021] [Indexed: 12/17/2022]
Abstract
Objectives Administration of medications such as dexmedetomidine as a topical anesthetic has been suggested in the pain control in dentistry. This double-blind randomized control trial study evaluated postoperative pain and associated factors following impacted third molar extraction surgery. Lidocaine alone was taken as the control and lidocaine plus dexmedetomidine as the intervention. Materials and Methods Forty patients undergoing mandibular third molar extraction entered the study and were randomly allocated to the control and interventional groups. 0.15 ml of dexmedetomidine was added to each lidocaine cartridge and the drug concentration was adjusted to 15 μg for the intervention group while only lidocaine was used in the control group. A visual analog scale was used to measure and record pain levels at the end of the surgery and 6, 12, and 24 hours after the surgery and number of painkillers taken by the patients after the surgery was also recorded. Results Pain scores of the intervention group decreased significantly during the surgery and also 6, 12, and 24 hours after the surgery compared to the control group. The pain score was correlated significantly with our intervention during the surgery and also 6 and 12 hours after that (all P value < 0.05). There was a nonsignificant reduction in the number of painkillers taken by the patients at 6, 12, and 24 hours after surgery (all P value > 0.05). Conclusion In patients undergoing molar surgery, administration of a combination of dexmedetomidine and lidocaine is beneficial for the pain control. Clinical Relevance. Compared to the injection of lidocaine alone, combination of dexmedetomidine and lidocaine can be used for a better pain control in molar surgeries.
Collapse
|
189
|
Zaouter C, Damphousse R, Moore A, Stevens LM, Gauthier A, Carrier FM. Elements not Graded in the Cardiac Enhanced Recovery After Surgery Guidelines Might Improve Postoperative Outcome: A Comprehensive Narrative Review. J Cardiothorac Vasc Anesth 2021; 36:746-765. [PMID: 33589344 DOI: 10.1053/j.jvca.2021.01.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022]
Abstract
Enhanced Recovery Programs (ERPs) are protocols involving the whole patient surgical journey. These protocols are based on multimodal, multidisciplinary, evidence-based, and patient-centered approaches aimed at improving patient recovery after a surgical intervention. Such programs have shown striking positive results in different surgical specialties. However, only a few research groups have incorporated preoperative, intraoperative, and postoperative evidence-based interventions in bundles used to standardize care and build cardiac surgery ERPs. The Enhanced Recovery After Surgery Society recently published evidence-based recommendations for perioperative care in cardiac surgery. Their recommendations included 22 perioperative interventions that may be part of any cardiac ERP. However, various components integrated in already-published cardiac ERPs were neither graded nor reported in these recommendations. The goals of the current review are to present published cardiac ERPs and their effects on patient outcomes and reported components incorporated into these ERPs and to discuss the objectives and scope of cardiac ERPs.
Collapse
Affiliation(s)
- Cédrick Zaouter
- Department of Anesthesiology, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada.
| | - Remy Damphousse
- Department of Anesthesiology, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Alex Moore
- Department of Anesthesiology, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Louis-Mathieu Stevens
- Department of Surgery, Division of Cardiac surgery, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Alain Gauthier
- Department of Anesthesiology, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - François Martin Carrier
- Department of Anesthesiology, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada; Department of Medicine, Division of Critical Care, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
190
|
Mendonça MM, Costa AN, Moraes GCA, Martins GM, Almeida AF, Rincon GCN, Siqueira JPR, Padilha DM, Moya MI, Ferreira-Neto ML, Gomes RM, Pedrino GR, Fontes MAP, Colombari E, Crestani CC, Fajemiroye JO, Xavier CH. Centrally acting antihypertensives change the psychogenic cardiovascular reactivity. Fundam Clin Pharmacol 2021; 35:892-905. [PMID: 33465820 DOI: 10.1111/fcp.12648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/21/2020] [Accepted: 01/15/2021] [Indexed: 11/27/2022]
Abstract
Clonidine (CL) and Rilmenidine (RI) are among the most frequently prescribed centrally acting antihypertensives. Here, we compared CL and RI effects on psychogenic cardiovascular reactivity to sonant, luminous, motosensory, and vibrotactile stimuli during neurogenic hypertension. The femoral artery and vein of Wistar (WT - normotensive) and spontaneously hypertensive rats (SHR) were catheterized before (24 h interval) i.p. injection of vehicle (NaCl 0.9%, control - CT group), CL (10 µg/kg), or RI (10 µg/kg) and acute exposure to luminous (5000 lm), sonant (75 dB sudden tap), motor (180° cage twist), and air-jet (10 L/min - restraint and vibrotactile). Findings showed that: (i) CL or RI reduced the arterial pressure of SHR, without affecting basal heart rate in WT and SHR; (ii) different stimuli evoked pressor and tachycardic responses; (iii) CL and RI reduced pressor response to sound; (iv) CL or RI reduced pressor responses to luminous stimulus without a change in peak tachycardia in SHR; (v) cage twist increased blood pressure in SHR, which was attenuated by CL or RI; (vi) air-jet increased pressure and heart rate; (vii) CL or RI attenuated the pressor responses to air-jet in SHR while RI reduced the chronotropic reactivity in both strains. Altogether, both antihypertensives relieved the psychogenic cardiovascular responses to different stimuli. The RI elicited higher cardioprotective effects through a reduction in air-jet-induced tachycardia.
Collapse
Affiliation(s)
- Michelle M Mendonça
- Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil
| | - Amanda N Costa
- Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil
| | - Gean C A Moraes
- Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil
| | - Gustavo M Martins
- Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil.,School of Medicine, Federal University of Goias, Goiania, Brazil
| | - Aline F Almeida
- Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil.,School of Medicine, Federal University of Goias, Goiania, Brazil
| | - Gabriel C N Rincon
- Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil.,School of Medicine, Federal University of Goias, Goiania, Brazil
| | - João P R Siqueira
- Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil.,School of Medicine, Federal University of Goias, Goiania, Brazil
| | - Daniella M Padilha
- Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil.,School of Medicine, Federal University of Goias, Goiania, Brazil
| | - Marcela I Moya
- Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil.,School of Medicine, Federal University of Goias, Goiania, Brazil
| | | | - Rodrigo Mello Gomes
- Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil
| | | | | | - Eduardo Colombari
- School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Carlos C Crestani
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - James O Fajemiroye
- Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil
| | | |
Collapse
|
191
|
Kobos L, Shannahan J. Particulate matter inhalation and the exacerbation of cardiopulmonary toxicity due to metabolic disease. Exp Biol Med (Maywood) 2021; 246:822-834. [PMID: 33467887 DOI: 10.1177/1535370220983275] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Particulate matter is a significant public health issue in the United States and globally. Inhalation of particulate matter is associated with a number of systemic and organ-specific adverse health outcomes, with the pulmonary and cardiovascular systems being particularly vulnerable. Certain subpopulations are well-recognized as being more susceptible to inhalation exposures, such as the elderly and those with pre-existing respiratory disease. Metabolic syndrome is becoming increasingly prevalent in our society and has known adverse effects on the heart, lungs, and vascular systems. The limited evaluations of individuals with metabolic syndromehave demonstrated that theymay compose a sensitive subpopulation to particulate exposures. However, the toxicological mechanisms responsible for this increased vulnerability are not fully understood. This review evaluates the currently available literature regarding how the response of an individual's pulmonary and cardiovascular systems is influenced by metabolic syndrome and metabolic syndrome-associated conditions such as hypertension, dyslipidemia, and diabetes. Further, we will discuss potential therapeutic agents and targets for the alleviation and treatment of particulate-matter induced metabolic illness. The information reviewed here may contribute to the understanding of metabolic illness as a risk factor for particulate matter exposure and further the development of therapeutic approaches to treat vulnerable subpopulations, such as those with metabolic diseases.
Collapse
Affiliation(s)
- Lisa Kobos
- School of Health Sciences, College of Human and Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jonathan Shannahan
- School of Health Sciences, College of Human and Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
192
|
Pepose JS, Hartman PJ, DuBiner HB, Abrams MA, Smyth-Medina RJ, Moroi SE, Meyer AR, Sooch MP, Jaber RM, Charizanis K, Klapman SA, Amin AT, Yousif JE, Lazar ES, Karpecki PM, Slonim CB, McDonald MB. Phentolamine Mesylate Ophthalmic Solution Provides Lasting Pupil Modulation and Improves Near Visual Acuity in Presbyopic Glaucoma Patients in a Randomized Phase 2b Clinical Trial. Clin Ophthalmol 2021; 15:79-91. [PMID: 33447013 PMCID: PMC7802916 DOI: 10.2147/opth.s278169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 12/04/2020] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Phentolamine mesylate ophthalmic solution (PMOS), applied to the eye topically, was shown previously to have beneficial effects in patients with dim light vision disturbances (DLD), including decreased pupil diameter (PD), improved best-corrected distance visual acuity (BCDVA), as well as lower intraocular pressure (IOP). The ORION-1 trial evaluated the long-term safety and efficacy of PMOS in a glaucomatous, presbyopic population. PATIENTS AND METHODS In this randomized, double-masked, multi-center, placebo-controlled, multiple-dose Phase 2b trial, 39 patients with elevated IOP were randomized to receive one evening dose of study medication or placebo for 14 days. The primary outcome measure was mean change in diurnal IOP, and the key secondary outcome measures included changes in PD, distance-corrected near visual acuity (DCNVA), and conjunctival hyperemia. RESULTS Use of 1% PMOS did not lead to a statistically significant decrease in diurnal IOP compared to placebo (P = 0.89) but trended toward a greater decrease in patients with lower IOP baselines. PMOS produced a statistically significant mean 20% PD reduction under both photopic and mesopic conditions that was sustained for 36 hours post-dosing. A statistically significant number of patients with PMOS compared to placebo demonstrated ≥1 line of improvement in photopic DCNVA at day 8 (P = 0.0018), day 15 (P = 0.0072), and day 16 (P = 0.0163), with a trend for 2- and 3-line improvements at all time points. There was no statistical difference in conjunctival hyperemia compared to placebo. CONCLUSION Although mean IOP was not lowered significantly, daily evening dosing of 1% PMOS was found to be well tolerated with no daytime conjunctival redness and demonstrated improvement in DCNVA with sustained PD reduction in a glaucomatous and presbyopic population. Smaller pupil size can have beneficial effects in improving symptoms of presbyopia and DLD, which will be the focus of further studies.
Collapse
Affiliation(s)
- Jay S Pepose
- Pepose Vision Institute, St. Louis, MO, USA
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Paul J Hartman
- Department of Ophthalmology, University of Rochester, Rochester, NY, USA
| | | | | | | | - Sayoko E Moroi
- The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | | | | | | | | | - Arin T Amin
- Ocuphire Pharma Inc, Farmington Hills, MI, USA
| | | | | | - Paul M Karpecki
- Kentucky College of Optometry, University of Pikeville, Pikeville, KY, USA
| | | | - Marguerite B McDonald
- Department of Ophthalmology, New York University Langone Medical Center, New York, NY, USA
| |
Collapse
|
193
|
Schultz KJ, Colby SM, Lin VS, Wright AT, Renslow RS. Ligand- and Structure-Based Analysis of Deep Learning-Generated Potential α2a Adrenoceptor Agonists. J Chem Inf Model 2021; 61:481-492. [PMID: 33404240 DOI: 10.1021/acs.jcim.0c01019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The α2a adrenoceptor is a medically relevant subtype of the G protein-coupled receptor family. Unfortunately, high-throughput techniques aimed at producing novel drug leads for this receptor have been largely unsuccessful because of the complex pharmacology of adrenergic receptors. As such, cutting-edge in silico ligand- and structure-based assessment and de novo deep learning methods are well positioned to provide new insights into protein-ligand interactions and potential active compounds. In this work, we (i) collect a dataset of α2a adrenoceptor agonists and provide it as a resource for the drug design community; (ii) use the dataset as a basis to generate candidate-active structures via deep learning; and (iii) apply computational ligand- and structure-based analysis techniques to gain new insights into α2a adrenoceptor agonists and assess the quality of the computer-generated compounds. We further describe how such assessment techniques can be applied to putative chemical probes with a case study involving proposed medetomidine-based probes.
Collapse
Affiliation(s)
- Katherine J Schultz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Sean M Colby
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Vivian S Lin
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Aaron T Wright
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States.,The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington 99163, United States
| | - Ryan S Renslow
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States.,The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington 99163, United States
| |
Collapse
|
194
|
Damian DB, Ghiță AM, Istrate S, Coman IC. Experimental research in rats on the reactivity of new corneal blood vessels to adrenaline. Rom J Ophthalmol 2021; 65:64-69. [PMID: 33817436 PMCID: PMC7995500 DOI: 10.22336/rjo.2021.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2021] [Indexed: 11/18/2022] Open
Abstract
Aim: The purpose of this experimental study was to evaluate the existence of adrenergic receptors in ketamine-induced corneal blood vessels in rat pups. Methods: The study of corneal neovascularization motricity was performed on 45-day-old Wistar rats in which, starting from the 15th day of life, corneal blood vessels were obtained by injecting intraperitoneal ketamine at a dose of 150 mg/ kg body weight, a total of 5 successive doses. The examination of the neovascularization was done with the help of a Nikon stereomicroscope connected to a video camera and a computer, the total magnification being 400X. The reactivity of the new corneal blood vessels to the administration in conjunctival instillations of a 1.5 mmol/L adrenaline solution was tested. The parameters followed were represented by variations in the caliber of corneal blood vessels. The data were analyzed using Microsoft Office Excel. Results: Administration of distilled water did not produce statistically significant changes in corneal blood vessels, while adrenaline produced a statistically significant constriction of vascular diameter (p=0.01 at T9, p=0.004 at T10, p=0.019 at time T11 of examinations). Conclusions: The results showed that adrenaline produces vasoconstriction in the new corneal blood vessels, which allows us to assume that they contain α-adrenergic receptors. However, we cannot say that corneal pathological vessels do not contain β2-type adrenergic receptors, because the effect of adrenaline may be an algebraic sum between vasoconstriction produced by stimulating α-adrenergic receptors and vasodilation produced by stimulating β2-adrenergic receptors, but in which the vasodilating effect may be masked by the vasoconstrictor effect given by a higher density of α-adrenergic receptors. Abbreviations: A= adrenaline, DNM = non-measurable diameter, NA= noradrenaline, Std.Er.= Standard error.
Collapse
Affiliation(s)
- Daniela Bianca Damian
- Department of Ophthalmology, “Dr. Alexandru Popescu” Military Emergency Hospital Focșani, Focșani, Vrancea, Romania
| | - Aurelian Mihai Ghiță
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Sânziana Istrate
- Department of Ophthalmology, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Ioana Cristina Coman
- Department of Ophthalmology, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
195
|
Glaess SS, Attridge RL, Christina Gutierrez G. Clonidine as a strategy for discontinuing dexmedetomidine sedation in critically ill patients: A narrative review. Am J Health Syst Pharm 2020; 77:515-522. [PMID: 32086509 DOI: 10.1093/ajhp/zxaa013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
PURPOSE To review the efficacy and safety of transitioning from dexmedetomidine to clonidine to facilitate weaning of patients from sedation with dexmedetomidine. There is a paucity of data describing dexmedetomidine withdrawal syndrome (DWS) as well as clonidine's place in therapy for DWS. This review will describe and analyze current literature to provide clinical recommendations. SUMMARY A MEDLINE literature search was performed to identify original research articles describing DWS and/or transitioning from dexmedetomidine to clonidine for the purpose of weaning patients from sedation with dexmedetomidine. Four case reports describing DWS, 3 case reports describing the use of clonidine to treat DWS, and 3 observational studies describing the use of clonidine to facilitate dexmedetomidine weaning were identified. The incidence of and risk factors for DWS are unknown; factors including patient age and dexmedetomidine infusion rate, loading dose, and discontinuation strategy have inconsistent associations with DWS. All cases of DWS have been associated with infusion durations greater than 72 hours. While there are limited data describing clonidine use for the treatment of dexmedetomidine withdrawal, clonidine appears to be beneficial for dexmedetomidine weaning and its use for that purpose has been well described. Clonidine dosages that have been assessed for discontinuing dexmedetomidine vary from 0.1 to 0.3 mg orally or enterally every 6 to 8 hours; one study assessed use of transdermal clonidine (100 µg/24 h patch). Patients with extensive cardiac comorbidities may be more susceptible to adverse effects of clonidine, which may limit the drug's use for DWS intervention. CONCLUSION Despite limited supportive data, clonidine provides a promising option for sedation management in adult ICU patients, with successful transitions from dexmedetomidine reported within 24 hours after clonidine initiation.
Collapse
Affiliation(s)
- Shelley S Glaess
- University of the Incarnate Word Feik School of Pharmacy, San Antonio, TX, and UCHealth Memorial Hospital, Colorado Springs, CO
| | - Rebecca L Attridge
- University of the Incarnate Word Feik School of Pharmacy, San Antonio, TX, and UT Health San Antonio, San Antonio, TX
| | - G Christina Gutierrez
- UT Health San Antonio, San Antonio, TX, and University Health System, San Antonio, TX
| |
Collapse
|
196
|
Windsor RB, Sierra M, Zappitelli M, McDaniel M. Beyond Amitriptyline: A Pediatric and Adolescent Oriented Narrative Review of the Analgesic Properties of Psychotropic Medications for the Treatment of Complex Pain and Headache Disorders. CHILDREN-BASEL 2020; 7:children7120268. [PMID: 33276542 PMCID: PMC7761583 DOI: 10.3390/children7120268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022]
Abstract
Children and adolescents with recurrent or chronic pain and headache are a complex and heterogenous population. Patients are best served by multi-specialty, multidisciplinary teams to assess and create tailored, individualized pain treatment and rehabilitation plans. Due to the complex nature of pain, generalizing pharmacologic treatment recommendations in children with recurrent or chronic pains is challenging. This is particularly true of complicated patients with co-existing painful and psychiatric conditions. There is an unfortunate dearth of evidence to support many pharmacologic therapies to treat children with chronic pain and headache. This narrative review hopes to supplement the available treatment options for this complex population by reviewing the pediatric and adult literature for analgesic properties of medications that also have psychiatric indication. The medications reviewed belong to medication classes typically described as antidepressants, alpha 2 delta ligands, mood stabilizers, anti-psychotics, anti-sympathetic agents, and stimulants.
Collapse
Affiliation(s)
- Robert Blake Windsor
- Division of Pediatric Pain Medicine, Department of Pediatrics, Prisma Health, Greenville, SC 29607, USA;
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29607, USA; (M.S.); (M.Z.)
- Correspondence:
| | - Michael Sierra
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29607, USA; (M.S.); (M.Z.)
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Prisma Health, Greenville, SC 29607, USA
| | - Megan Zappitelli
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29607, USA; (M.S.); (M.Z.)
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Prisma Health, Greenville, SC 29607, USA
| | - Maria McDaniel
- Division of Pediatric Pain Medicine, Department of Pediatrics, Prisma Health, Greenville, SC 29607, USA;
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29607, USA; (M.S.); (M.Z.)
| |
Collapse
|
197
|
Jiang S, Czuma R, Cohen-Oram A, Hartney K, Stern TA. Guanfacine for Hyperactive Delirium: A Case Series. J Acad Consult Liaison Psychiatry 2020; 62:83-88. [PMID: 33272699 DOI: 10.1016/j.psym.2020.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 11/25/2022]
Affiliation(s)
- Shixie Jiang
- Department of Psychiatry and Behavioral Neurosciences, Tampa General Hospital, University of South Florida, Tampa, FL.
| | - Richard Czuma
- Department of Psychiatry and Behavioral Neurosciences, Tampa General Hospital, University of South Florida, Tampa, FL
| | - Alexis Cohen-Oram
- Department of Psychiatry and Behavioral Neurosciences, Tampa General Hospital, University of South Florida, Tampa, FL
| | - Kimberly Hartney
- Department of Psychiatry and Behavioral Neurosciences, Tampa General Hospital, University of South Florida, Tampa, FL
| | - Theodore A Stern
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
198
|
Lisowska B, Jakubiak J, Siewruk K, Sady M, Kosson D. Which idea is better with regard to immune response? Opioid anesthesia or opioid free anesthesia. J Inflamm Res 2020; 13:859-869. [PMID: 33177861 PMCID: PMC7652233 DOI: 10.2147/jir.s275986] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
The stress of surgery is characterized by an inflammatory response with immune suppression resulting from many factors, including the type of surgery and the kind of anesthesia, linked with the drugs that are used and the underlying disease of the patient. The trauma of surgery triggers a cascade of reactions involving the immune response and nociception. As strong analgesics, opioids provide the analgesic component of general anesthesia with bi-directional effect on the immune system. Opioids influence almost all aspects of the immune response in regards to leukocytes, macrophages, mast cells, lymphocytes, and NK cells. The suppressive effect of opioids on the immune system is limiting their use, especially in patients with impaired immune response, so the possibility of using multimodal anesthesia without opioids, known as opioid-free anesthesia (OFA), is gaining more and more sympathizers. The idea of OFA is to eliminate opioid analgesia in the treatment of acute pain and to replace it with drugs from other groups that are assumed to have a comparable analgesic effect without affecting the immune system. Here, we present a review on the impact of anesthesia, with and without the use of opioids, on the immune response to surgical stress.
Collapse
Affiliation(s)
- Barbara Lisowska
- Department Anesthesiology and Intensive Medical Care, National Geriatrics, Rheumatology and Rehabilitation Institute, Warsaw 02-637, Poland
| | - Jakub Jakubiak
- Department of Anesthesiology and Intensive Care, John Paul II Western Hospital, Grodzisk Mazowiecki 05-825, Poland
| | - Katarzyna Siewruk
- Faculty of Veterinary Medicine, Department of Large Animal Diseases with Clinic, Warsaw University of Life Sciences, Warsaw 02-797, Poland
| | - Maria Sady
- Faculty of Veterinary Medicine, Department of Large Animal Diseases with Clinic, Warsaw University of Life Sciences, Warsaw 02-797, Poland
| | - Dariusz Kosson
- Department of Anaesthesiology and Intensive Care, Division of Teaching, Medical University of Warsaw, Warsaw 02-005, Poland
| |
Collapse
|
199
|
Pathan S, Kaplan JB, Adamczyk K, Chiu SH, Shah CV. Evaluation of dexmedetomidine withdrawal in critically ill adults. J Crit Care 2020; 62:19-24. [PMID: 33227592 DOI: 10.1016/j.jcrc.2020.10.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND Dexmedetomidine (DEX) withdrawal syndrome has been reported in the pediatric population, but literature describing DEX withdrawal in critically ill adults is limited. The purpose of this study was to determine the incidence of DEX withdrawal in adult patients and to identify factors associated with DEX withdrawal syndrome. METHODS A retrospective chart review was performed in the adult intensive care units of two tertiary medical centers. Eligible patients were at least 18 years of age and received DEX for 24 h or more. Patients were excluded if they presented with a primary neurologic diagnosis, had a history of substance abuse, or received any other α2-agonists 24 h before discontinuation of DEX. The primary outcome was the percentage of patients who developed withdrawal as defined by the presence of two or more symptoms (tachycardia, hypertension, vomiting, agitation) within the 24 h following DEX discontinuation. RESULTS Of the 165 patients included, 50 patients experienced withdrawal (30.3%), lasting a median of two days. The incidence of withdrawal was higher in surgical (40%) compared to medical (28%) or cardiac (32%) patients (p = 0.004). Median duration of infusion was 52.5 h (interquartile range [IQR], 37.8 to 102.8) in the withdrawal group and 52 h (IQR, 41 to 87) in the non-withdrawal group (p = 0.887). Median DEX dose was 0.56 μg/kg/h (IQR, 0.39 to 0.83) in the withdrawal group and 0.48 μg/kg/h (0.36 to 0.65) in the non-withdrawal group (p = 0.12). Weaning did not reduce the incidence of withdrawal as compared to abrupt discontinuation (p = 0.68). The withdrawal group was more likely to have concomitantly discontinued opioids (54% vs 12.2%) and benzodiazepines (36% vs 0%) at the time of DEX discontinuation compared to the non-withdrawal group (p = 0.004). CONCLUSION Development of DEX-associated withdrawal occurred in approximately 30% of adult patients, comparable to rates reported in pediatric literature. There appeared to be no correlation between dose, exposure, and weaning in the occurrence of withdrawal, but concomitant discontinuation of opioids or benzodiazepines as well as ICU admission type could highlight cases requiring closer monitoring.
Collapse
Affiliation(s)
- Sophia Pathan
- Department of Pharmacy, The Johns Hopkins Hospital, United States of America.
| | - Justin B Kaplan
- Department of Pharmacy, Atlantic Health System, United States of America
| | - Katarzyna Adamczyk
- Department of Pharmacy, Atlantic Health System, United States of America
| | - Stephanie H Chiu
- Atlantic Center for Research, Atlantic Health System, United States of America
| | - Chirag V Shah
- Department of Medicine, Atlantic Health System, United States of America
| |
Collapse
|
200
|
Zhu H, Ren A, Zhou K, Chen Q, Zhang M, Liu J. Impact of Dexmedetomidine Infusion on Postoperative Acute Kidney Injury in Elderly Patients Undergoing Major Joint Replacement: A Retrospective Cohort Study. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4695-4701. [PMID: 33173279 PMCID: PMC7646437 DOI: 10.2147/dddt.s278342] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Purpose Postoperative acute kidney injury (AKI) is a frequent complication in elderly patients that increases morbidity and mortality. Approximately 1.7 million people die from AKI worldwide every year. Dexmedetomidine (Dex) is often used as an adjunct to multimodal analgesia. Our study investigated whether Dex could safely decrease the incidence of AKI in elderly patients undergoing major joint replacement. Methods A single-center retrospective study was conducted in patients aged >65 years undergoing major joint replacement. Propensity score–matching analysis was used, and a total of 1,006 patients were matched successfully. The primary outcome was the incidence of postoperative AKI. Secondary outcomes included perioperative adverse complications, opioid consumption, time to extubation, and length of hospital stay. Results Among the 1,006 patients included, postoperative AKI occurred in 9.3% (n=94). The Dex group (perioperative Dex infusion) had lower incidence of postoperative AKI than the control group (7.2% vs 11.5%, P=0.017). Compared with the control group, the Dex group had less opioid consumption (P<0.05), reduced time to extubation (P=0.004), and shorter length of hospital stay (P=0.001). The Dex group also showed higher incidence of bradycardia (20.1% vs 15.1%, P=0.038). There were no differences in intraoperative hypotension (19.5% vs 17.5%), postoperative nausea and vomiting (4.2% vs 5.4%), time in PACU (45.0±6.4 vs 45.5±6.2 minutes), or rate of ICU admission (9.7% vs 11.1%) between the Dex group and control group (All P>0.05). Conclusion This retrospective study showed Dex infusion in elderly patients undergoing major joint replacement was associated with lower incidence of postoperative AKI, less opioid consumption, and shorter extubation time and hospital stay. However, the Dex group had higher incidence of bradycardia. We found no statistical differences in other perioperative adverse complications between the groups.
Collapse
Affiliation(s)
- He Zhu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Aolin Ren
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Kang Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Qiuchong Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Mengjun Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Jindong Liu
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| |
Collapse
|