51
|
Chou T, Nabavinia M, Tram NK, Rimmerman ET, Patel S, Musini KN, Eisert SN, Wolfe T, Wynveen MK, Matsuzaki Y, Kitsuka T, Iwaki R, Janse SA, Bobbey AJ, Breuer CK, Goodchild L, Malbrue R, Shinoka T, Atway SA, Go MR, Stacy MR. Quantification of Skeletal Muscle Perfusion in Peripheral Artery Disease Using 18F-Sodium Fluoride Positron Emission Tomography Imaging. J Am Heart Assoc 2024; 13:e031823. [PMID: 38353265 PMCID: PMC11010069 DOI: 10.1161/jaha.123.031823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/07/2023] [Indexed: 02/16/2024]
Abstract
BACKGROUND Perfusion deficits contribute to symptom severity, morbidity, and death in peripheral artery disease (PAD); however, no standard method for quantifying absolute measures of skeletal muscle perfusion exists. This study sought to preclinically test and clinically translate a positron emission tomography (PET) imaging approach using an atherosclerosis-targeted radionuclide, fluorine-18-sodium fluoride (18F-NaF), to quantify absolute perfusion in PAD. METHODS AND RESULTS Eight Yorkshire pigs underwent unilateral femoral artery ligation and dynamic 18F-NaF PET/computed tomography imaging on the day of and 2 weeks after occlusion. Following 2-week imaging, calf muscles were harvested to quantify microvascular density. PET methodology was validated with microspheres in 4 additional pig studies and translated to patients with PAD (n=39) to quantify differences in calf perfusion across clinical symptoms/stages and perfusion responses in a case of revascularization. Associations between PET perfusion, ankle-brachial index, toe-brachial index, and toe pressure were assessed in relation to symptoms. 18F-NaF PET/computed tomography quantified significant deficits in calf perfusion in pigs following arterial occlusion and perfusion recovery 2 weeks after occlusion that coincided with increased muscle microvascular density. Additional studies confirmed that PET-derived perfusion measures agreed with microsphere-derived perfusion measures. Translation of imaging methods demonstrated significant decreases in calf perfusion with increasing severity of PAD and quantified perfusion responses to revascularization. Perfusion measures were also significantly associated with symptom severity, whereas traditional hemodynamic measures were not. CONCLUSIONS 18F-NaF PET imaging quantifies perfusion deficits that correspond to clinical stages of PAD and represents a novel perfusion imaging strategy that could be partnered with atherosclerosis-targeted 18F-NaF PET imaging using a single radioisotope injection. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT03622359.
Collapse
Affiliation(s)
- Ting‐Heng Chou
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Mahboubeh Nabavinia
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Nguyen K. Tram
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Eleanor T. Rimmerman
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
- Biophysics Graduate ProgramOhio State UniversityColumbusOH
| | - Surina Patel
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Kumudha Narayana Musini
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Susan Natalie Eisert
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Tatiana Wolfe
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Molly K. Wynveen
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Yuichi Matsuzaki
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Takahiro Kitsuka
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Ryuma Iwaki
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | | | - Adam J. Bobbey
- Department of RadiologyNationwide Children’s HospitalColumbusOH
| | - Christopher K. Breuer
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Laurie Goodchild
- Animal Resources CoreResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Raphael Malbrue
- Animal Resources CoreResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Toshiharu Shinoka
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
| | - Said A. Atway
- Department of OrthopaedicsOhio State University College of MedicineColumbusOH
| | - Michael R. Go
- Division of Vascular Diseases & Surgery, Department of SurgeryOhio State University College of MedicineColumbusOH
| | - Mitchel R. Stacy
- Center for Regenerative MedicineResearch Institute at Nationwide Children’s HospitalColumbusOH
- Biophysics Graduate ProgramOhio State UniversityColumbusOH
- Division of Vascular Diseases & Surgery, Department of SurgeryOhio State University College of MedicineColumbusOH
| |
Collapse
|
52
|
Cheng CJ, Nizar JM, Dai DF, Huang CL. Transport activity regulates mitochondrial bioenergetics and biogenesis in renal tubules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.04.578838. [PMID: 38370657 PMCID: PMC10871199 DOI: 10.1101/2024.02.04.578838] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Renal tubules are featured with copious mitochondria and robust transport activity. Mutations in mitochondrial genes cause congenital renal tubulopathies, and changes in transport activity affect mitochondrial morphology, suggesting mitochondrial function and transport activity are tightly coupled. Current methods of using bulk kidney tissues or cultured cells to study mitochondrial bioenergetics are limited. Here, we optimized an extracellular flux analysis (EFA) to study mitochondrial respiration and energy metabolism using microdissected mouse renal tubule segments. EFA detects mitochondrial respiration and glycolysis by measuring oxygen consumption and extracellular acidification rates, respectively. We show that both measurements positively correlate with sample sizes of a few centimeter-length renal tubules. The thick ascending limbs (TALs) and distal convoluted tubules (DCTs) predominantly utilize glucose/pyruvate as energy substrates, whereas proximal tubules (PTs) are significantly much less so. Acute inhibition of TALs' transport activity by ouabain treatment reduces basal and ATP-linked mitochondrial respiration. Chronic inhibition of transport activity by 2-week furosemide treatment or deletion of with-no-lysine kinase 4 (Wnk4) decreases maximal mitochondrial capacity. In addition, chronic inhibition downregulates mitochondrial DNA mass and mitochondrial length/density in TALs and DCTs. Conversely, gain-of-function Wnk4 mutation increases maximal mitochondrial capacity and mitochondrial length/density without increasing mitochondrial DNA mass. In conclusion, EFA is a sensitive and reliable method to investigate mitochondrial functions in isolated renal tubules. Transport activity tightly regulates mitochondrial bioenergetics and biogenesis to meet the energy demand in renal tubules. The system allows future investigation into whether and how mitochondria contribute to tubular remodeling adapted to changes in transport activity. Key points A positive correlation between salt reabsorption and oxygen consumption in mammalian kidneys hints at a potential interaction between transport activity and mitochondrial respiration in renal tubules.Renal tubules are heterogeneous in transport activity and mitochondrial metabolism, and traditional assays using bulk kidney tissues cannot provide segment-specific information.Here, we applied an extracellular flux analysis to investigate mitochondrial respiration and energy metabolism in isolated renal tubules. This assay is sensitive in detecting oxygen consumption and acid production in centimeter-length renal tubules and reliably recapitulates segment-specific metabolic features.Acute inhibition of transport activity reduces basal and ATP-linked mitochondrial respirations without changing maximal mitochondrial respiratory capacity. Chronic alterations of transport activity further adjust maximal mitochondrial respiratory capacity via regulating mitochondrial biogenesis or non-transcriptional mechanisms.Our findings support the concept that renal tubular cells finely adjust mitochondrial bioenergetics and biogenesis to match the new steady state of transport activity.
Collapse
|
53
|
Wood G, Madsen TL, Kim WY, Lyhne MD. Increasing Levels of Positive End-expiratory Pressure Cause Stepwise Biventricular Stroke Work Reduction in a Porcine Model. Anesthesiology 2024; 140:240-250. [PMID: 37905995 DOI: 10.1097/aln.0000000000004821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
BACKGROUND Positive end-expiratory pressure (PEEP) is commonly applied to avoid atelectasis and improve oxygenation in patients during general anesthesia but affects cardiac pressures, volumes, and loading conditions through cardiorespiratory interactions. PEEP may therefore alter stroke work, which is the area enclosed by the pressure-volume loop and corresponds to the external work performed by the ventricles to eject blood. The low-pressure right ventricle may be even more susceptible to PEEP than the left ventricle. The authors hypothesized that increasing levels of PEEP would reduce stroke work in both ventricles. METHODS This was a prospective, observational, experimental study. Six healthy female pigs of approximately 60 kg were used. PEEP was stepwise increased from 0 to 5, 7, 9, 11, 13, 15, 17, and 20 cm H2O to cover the clinical spectrum of PEEP. Simultaneous, biventricular invasive pressure-volume loops, invasive blood pressures, and ventilator data were recorded. RESULTS Increasing PEEP resulted in stepwise reductions in left (5,740 ± 973 vs. 2,303 ± 1,154 mmHg · ml; P < 0.001) and right (2,064 ± 769 vs. 468 ± 133 mmHg · ml; P < 0.001) ventricular stroke work. The relative stroke work reduction was similar between the two ventricles. Left ventricular ejection fraction, afterload, and coupling were preserved. On the contrary, PEEP increased right ventricular afterload and caused right ventriculo-arterial uncoupling (0.74 ± 0.30 vs. 0.19 ± 0.13; P = 0.01) with right ventricular ejection fraction reduction (64 ± 8% vs. 37 ± 7%, P < 0.001). CONCLUSIONS A stepwise increase in PEEP caused stepwise reduction in biventricular stroke work. However, there are important interventricular differences in response to increased PEEP levels. PEEP increased right ventricular afterload leading to uncoupling and right ventricular ejection fraction decline. These findings may support clinical decision-making to further optimize PEEP as a means to balance between improving lung ventilation and preserving right ventricular function. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Gregory Wood
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Tobias Lynge Madsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Won Yong Kim
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Mads Dam Lyhne
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Anesthesiology and Intensive Care, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
54
|
Garcia-Bonilla L, Shahanoor Z, Sciortino R, Nazarzoda O, Racchumi G, Iadecola C, Anrather J. Analysis of brain and blood single-cell transcriptomics in acute and subacute phases after experimental stroke. Nat Immunol 2024; 25:357-370. [PMID: 38177281 DOI: 10.1038/s41590-023-01711-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 11/13/2023] [Indexed: 01/06/2024]
Abstract
Cerebral ischemia triggers a powerful inflammatory reaction involving peripheral leukocytes and brain resident cells that contribute to both tissue injury and repair. However, their dynamics and diversity remain poorly understood. To address these limitations, we performed a single-cell transcriptomic study of brain and blood cells 2 or 14 days after ischemic stroke in mice. We observed a strong divergence of post-ischemic microglia, monocyte-derived macrophages and neutrophils over time, while endothelial cells and brain-associated macrophages showed altered transcriptomic signatures at 2 days poststroke. Trajectory inference predicted the in situ trans-differentiation of macrophages from blood monocytes into day 2 and day 14 phenotypes, while neutrophils were projected to be continuously de novo recruited from the blood. Brain single-cell transcriptomes from both female and male aged mice were similar to that of young male mice, but aged and young brains differed in their immune cell composition. Although blood leukocyte analysis also revealed altered transcriptomes after stroke, brain-infiltrating leukocytes displayed higher transcriptomic divergence than their circulating counterparts, indicating that phenotypic diversification occurs within the brain in the early and recovery phases of ischemic stroke. A portal ( https://anratherlab.shinyapps.io/strokevis/ ) is provided to allow user-friendly access to our data.
Collapse
Affiliation(s)
- Lidia Garcia-Bonilla
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Ziasmin Shahanoor
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Rose Sciortino
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Omina Nazarzoda
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Gianfranco Racchumi
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Josef Anrather
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
55
|
Chen HS, van Roon L, Ge Y, van Gils JM, Schoones JW, DeRuiter MC, Zeppenfeld K, Jongbloed MRM. The relevance of the superior cervical ganglion for cardiac autonomic innervation in health and disease: a systematic review. Clin Auton Res 2024; 34:45-77. [PMID: 38393672 PMCID: PMC10944423 DOI: 10.1007/s10286-024-01019-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/22/2024] [Indexed: 02/25/2024]
Abstract
PURPOSE The heart receives cervical and thoracic sympathetic contributions. Although the stellate ganglion is considered the main contributor to cardiac sympathetic innervation, the superior cervical ganglia (SCG) is used in many experimental studies. The clinical relevance of the SCG to cardiac innervation is controversial. We investigated current morphological and functional evidence as well as controversies on the contribution of the SCG to cardiac innervation. METHODS A systematic literature review was conducted in PubMed, Embase, Web of Science, and COCHRANE Library. Included studies received a full/text review and quality appraisal. RESULTS Seventy-six eligible studies performed between 1976 and 2023 were identified. In all species studied, morphological evidence of direct or indirect SCG contribution to cardiac innervation was found, but its contribution was limited. Morphologically, SCG sidedness may be relevant. There is indirect functional evidence that the SCG contributes to cardiac innervation as shown by its involvement in sympathetic overdrive reactions in cardiac disease states. A direct functional contribution was not found. Functional data on SCG sidedness was largely unavailable. Information about sex differences and pre- and postnatal differences was lacking. CONCLUSION Current literature mainly supports an indirect involvement of the SCG in cardiac innervation, via other structures and plexuses or via sympathetic overdrive in response to cardiac diseases. Morphological evidence of a direct involvement was found, but its contribution seems limited. The relevance of SCG sidedness, sex, and developmental stage in health and disease remains unclear and warrants further exploration.
Collapse
Affiliation(s)
- H Sophia Chen
- Department of Cardiology, Willem Einthoven Center for Cardiac Arrhythmia Research and Management, Leiden University Medical Center, Leiden, The Netherlands
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lieke van Roon
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yang Ge
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Janine M van Gils
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan W Schoones
- Directorate of Research Policy, Leiden University Medical Center, Leiden, The Netherlands
| | - Marco C DeRuiter
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Katja Zeppenfeld
- Department of Cardiology, Willem Einthoven Center for Cardiac Arrhythmia Research and Management, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, Center of Congenital Heart Disease Amsterdam Leiden (CAHAL), Leiden University Medical Center, Leiden, The Netherlands
| | - Monique R M Jongbloed
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands.
- Department of Cardiology, Center of Congenital Heart Disease Amsterdam Leiden (CAHAL), Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
56
|
Geiseler SJ, Hadzic A, Lambertus M, Forbord KM, Sajedi G, Liesz A, Morland C. L-Lactate Treatment at 24 h and 48 h after Acute Experimental Stroke Is Neuroprotective via Activation of the L-Lactate Receptor HCA 1. Int J Mol Sci 2024; 25:1232. [PMID: 38279234 PMCID: PMC10816130 DOI: 10.3390/ijms25021232] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
Stroke is the main cause for acquired disabilities. Pharmaceutical or mechanical removal of the thrombus is the cornerstone of stroke treatment but can only be administered to a subset of patients and within a narrow time window. Novel treatment options are therefore required. Here we induced stroke by permanent occlusion of the distal medial cerebral artery of wild-type mice and knockout mice for the lactate receptor hydroxycarboxylic acid receptor 1 (HCA1). At 24 h and 48 h after stroke induction, we injected L-lactate intraperitoneal. The resulting atrophy was measured in Nissl-stained brain sections, and capillary density and neurogenesis were measured after immunolabeling and confocal imaging. In wild-type mice, L-lactate treatment resulted in an HCA1-dependent reduction in the lesion volume accompanied by enhanced angiogenesis. In HCA1 knockout mice, on the other hand, there was no increase in angiogenesis and no reduction in lesion volume in response to L-lactate treatment. Nevertheless, the lesion volumes in HCA1 knockout mice-regardless of L-lactate treatment-were smaller than in control mice, indicating a multifactorial role of HCA1 in stroke. Our findings suggest that L-lactate administered 24 h and 48 h after stroke is protective in stroke. This represents a time window where no effective treatment options are currently available.
Collapse
Affiliation(s)
- Samuel J. Geiseler
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, 0316 Oslo, Norway; (A.H.); (M.L.); (K.M.F.); (G.S.)
| | - Alena Hadzic
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, 0316 Oslo, Norway; (A.H.); (M.L.); (K.M.F.); (G.S.)
| | - Marvin Lambertus
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, 0316 Oslo, Norway; (A.H.); (M.L.); (K.M.F.); (G.S.)
| | - Karl Martin Forbord
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, 0316 Oslo, Norway; (A.H.); (M.L.); (K.M.F.); (G.S.)
| | - Ghazal Sajedi
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, 0316 Oslo, Norway; (A.H.); (M.L.); (K.M.F.); (G.S.)
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, 81377 Munich, Germany;
- Graduate School of Systemic Neurosciences Munich, 82152 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Cecilie Morland
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, 0316 Oslo, Norway; (A.H.); (M.L.); (K.M.F.); (G.S.)
| |
Collapse
|
57
|
Tran KKN, Wong VHY, Vessey KA, Finkelstein DI, Bui BV, Nguyen CTO. Levodopa Rescues Retinal Function in the Transgenic A53T Alpha-Synuclein Model of Parkinson's Disease. Biomedicines 2024; 12:130. [PMID: 38255235 PMCID: PMC10813165 DOI: 10.3390/biomedicines12010130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/11/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Loss of substantia nigra dopaminergic cells and alpha-synuclein (α-syn)-rich intraneuronal deposits within the central nervous system are key hallmarks of Parkinson's disease (PD). Levodopa (L-DOPA) is the current gold-standard treatment for PD. This study aimed to evaluate in vivo retinal changes in a transgenic PD model of α-syn overexpression and the effect of acute levodopa (L-DOPA) treatment. METHODS Anaesthetised 6-month-old mice expressing human A53T alpha-synuclein (HOM) and wildtype (WT) control littermates were intraperitoneally given 20 mg/kg L-DOPA (50 mg levodopa, 2.5 mg benserazide) or vehicle saline (n = 11-18 per group). In vivo retinal function (dark-adapted full-field ERG) and structure (optical coherence tomography, OCT) were recorded before and after drug treatment for 30 min. Ex vivo immunohistochemistry (IHC) on flat-mounted retina was conducted to assess tyrosine hydroxylase (TH) positive cell counts (n = 7-8 per group). RESULTS We found that photoreceptor (a-wave) and bipolar cell (b-wave) ERG responses (p < 0.01) in A53T HOM mice treated with L-DOPA grew in amplitude more (47 ± 9%) than WT mice (16 ± 9%) treated with L-DOPA, which was similar to the vehicle group (A53T HOM 25 ± 9%; WT 19 ± 7%). While outer retinal thinning (outer nuclear layer, ONL, and outer plexiform layer, OPL) was confirmed in A53T HOM mice (p < 0.01), L-DOPA did not have an ameliorative effect on retinal layer thickness. These findings were observed in the absence of changes to the number of TH-positive amacrine cells across experiment groups. Acute L-DOPA treatment transiently improves visual dysfunction caused by abnormal alpha-synuclein accumulation. CONCLUSIONS These findings deepen our understanding of dopamine and alpha-synuclein interactions in the retina and provide a high-throughput preclinical framework, primed for translation, through which novel therapeutic compounds can be objectively screened and assessed for fast-tracking PD drug discovery.
Collapse
Affiliation(s)
- Katie K. N. Tran
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC 3010, Australia; (K.K.N.T.); (V.H.Y.W.); (B.V.B.)
| | - Vickie H. Y. Wong
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC 3010, Australia; (K.K.N.T.); (V.H.Y.W.); (B.V.B.)
| | - Kirstan A. Vessey
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - David I. Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Bang V. Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC 3010, Australia; (K.K.N.T.); (V.H.Y.W.); (B.V.B.)
| | - Christine T. O. Nguyen
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC 3010, Australia; (K.K.N.T.); (V.H.Y.W.); (B.V.B.)
| |
Collapse
|
58
|
Lipp HP, Krackow S, Turkes E, Benner S, Endo T, Russig H. IntelliCage: the development and perspectives of a mouse- and user-friendly automated behavioral test system. Front Behav Neurosci 2024; 17:1270538. [PMID: 38235003 PMCID: PMC10793385 DOI: 10.3389/fnbeh.2023.1270538] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/18/2023] [Indexed: 01/19/2024] Open
Abstract
IntelliCage for mice is a rodent home-cage equipped with four corner structures harboring symmetrical double panels for operant conditioning at each of the two sides, either by reward (access to water) or by aversion (non-painful stimuli: air-puffs, LED lights). Corner visits, nose-pokes and actual licks at bottle-nipples are recorded individually using subcutaneously implanted transponders for RFID identification of up to 16 adult mice housed in the same home-cage. This allows for recording individual in-cage activity of mice and applying reward/punishment operant conditioning schemes in corners using workflows designed on a versatile graphic user interface. IntelliCage development had four roots: (i) dissatisfaction with standard approaches for analyzing mouse behavior, including standardization and reproducibility issues, (ii) response to handling and housing animal welfare issues, (iii) the increasing number of mouse models had produced a high work burden on classic manual behavioral phenotyping of single mice. and (iv), studies of transponder-chipped mice in outdoor settings revealed clear genetic behavioral differences in mouse models corresponding to those observed by classic testing in the laboratory. The latter observations were important for the development of home-cage testing in social groups, because they contradicted the traditional belief that animals must be tested under social isolation to prevent disturbance by other group members. The use of IntelliCages reduced indeed the amount of classic testing remarkably, while its flexibility was proved in a wide range of applications worldwide including transcontinental parallel testing. Essentially, two lines of testing emerged: sophisticated analysis of spontaneous behavior in the IntelliCage for screening of new genetic models, and hypothesis testing in many fields of behavioral neuroscience. Upcoming developments of the IntelliCage aim at improved stimulus presentation in the learning corners and videotracking of social interactions within the IntelliCage. Its main advantages are (i) that mice live in social context and are not stressfully handled for experiments, (ii) that studies are not restricted in time and can run in absence of humans, (iii) that it increases reproducibility of behavioral phenotyping worldwide, and (iv) that the industrial standardization of the cage permits retrospective data analysis with new statistical tools even after many years.
Collapse
Affiliation(s)
- Hans-Peter Lipp
- Faculty of Medicine, Institute of Evolutionary Medicine, University of Zürich, Zürich, Switzerland
| | - Sven Krackow
- Institute of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Emir Turkes
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Seico Benner
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Ibaraki, Japan
| | | | | |
Collapse
|
59
|
Ghasemi A, Jeddi S, Yousefzadeh N, Kashfi K, Norouzirad R. Dissolving sodium hydrosulfide in drinking water is not a good source of hydrogen sulfide for animal studies. Sci Rep 2023; 13:21839. [PMID: 38071388 PMCID: PMC10710449 DOI: 10.1038/s41598-023-49437-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023] Open
Abstract
Hydrogen sulfide (H2S) has many physiological and pathological roles in the human body. Sodium hydrosulfide (NaHS) is widely used as a pharmacological tool for assessing H2S effects in biological experiments. Although H2S loss from NaHS solution is a matter of minutes, some animal studies use NaHS in solution as an H2S-donating compound in drinking water. This study addresses whether 30 μM NaHS in drinking water prepared in rat/mouse water bottles remains stable for at least 12-24 h, as presumed by some authors. NaHS solutions (30 μM) were prepared in drinking water and immediately transferred to rat/mice water bottles. Samples were obtained from the tip of water bottles and from inside of the bottles at 0, 1, 2, 3, 4, 5, 6, 12, and 24 h for sulfide measurement using the methylene blue method. Furthermore, NaHS (30 μM) was administered to male and female rats for two weeks, and serum sulfide concentrations were measured every other day in the first week and at the end of the second week. NaHS solution was unstable in the samples obtained from the tip of water bottles; it declined by 72% and 75% after 12 and 24 h, respectively. In the samples obtained from the inside of the water bottles, the decline in the NaHS was not significant until 2 h; however, it decreased by 47% and 72% after 12 and 24 h, respectively. NaHS administration did not affect serum sulfide levels in male and female rats. In conclusion, NaHS solution prepared in drinking water can not be used for H2S donation as the solution is unstable. This route of administration exposes animals to variable and lower-than-expected amounts of NaHS.
Collapse
Affiliation(s)
- Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasibeh Yousefzadeh
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Reza Norouzirad
- Department of Biochemistry, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran.
| |
Collapse
|
60
|
Chen HS, van Roon L, Schoones J, Zeppenfeld K, DeRuiter MC, Jongbloed MRM. Cardiac sympathetic hyperinnervation after myocardial infarction: a systematic review and qualitative analysis. Ann Med 2023; 55:2283195. [PMID: 38065671 PMCID: PMC10836288 DOI: 10.1080/07853890.2023.2283195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Cardiac sympathetic hyperinnervation after myocardial infarction (MI) is associated with arrhythmogenesis and sudden cardiac death. The characteristics of cardiac sympathetic hyperinnervation remain underexposed. OBJECTIVE To provide a systematic review on cardiac sympathetic hyperinnervation after MI, taking into account: (1) definition, experimental model and quantification method and (2) location, amount and timing, in order to obtain an overview of current knowledge and to expose gaps in literature. METHODS References on cardiac sympathetic hyperinnervation were screened for inclusion. The included studies received a full-text review and quality appraisal. Relevant data on hyperinnervation were collected and qualitatively analysed. RESULTS Our literature search identified 60 eligible studies performed between 2000 and 2022. Cardiac hyperinnervation is generally defined as an increased sympathetic nerve density or increased number of nerves compared to another control group (100%). Studies were performed in a multitude of experimental models, but most commonly in male rats with permanent left anterior descending (LAD) artery ligation (male: 63%, rat: 68%, permanent ligation: 93%, LAD: 97%). Hyperinnervation seems to occur mainly in the borderzone. Quantification after MI was performed in regions of interest in µm2/mm2 (41%) or in percentage of nerve fibres (46%) and the reported amount showed a great variation ranging from 439 to 126,718 µm2/mm2. Hyperinnervation seems to start from three days onwards to >3 months without an evident peak, although studies on structural evaluation over time and in the chronic phase were scarce. CONCLUSIONS Cardiac sympathetic hyperinnervation after MI occurs mainly in the borderzone from three days onwards and remains present at later timepoints, for at least 3 months. It is most commonly studied in male rats with permanent LAD ligation. The amount of hyperinnervation differs greatly between studies, possibly due to differential quantification methods. Further studies are required that evaluate cardiac sympathetic hyperinnervation over time and in the chronic phase, in transmural sections, in the female sex, and in MI with reperfusion.
Collapse
Affiliation(s)
- H. Sophia Chen
- Department of Cardiology, Center of Congenital Heart Disease Amsterdam Leiden (CAHAL), Leiden University Medical Center, Leiden, The Netherlands
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lieke van Roon
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Schoones
- Dictorate of Research Policy, Leiden University Medical Center, Leiden, The Netherlands
| | - Katja Zeppenfeld
- Department of Cardiology, Center of Congenital Heart Disease Amsterdam Leiden (CAHAL), Leiden University Medical Center, Leiden, The Netherlands
| | - Marco C. DeRuiter
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique R. M. Jongbloed
- Department of Cardiology, Center of Congenital Heart Disease Amsterdam Leiden (CAHAL), Leiden University Medical Center, Leiden, The Netherlands
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
61
|
Huang Z, Peng Y, Ke G, Xiao Y, Chen Y. CaMKII may regulate renal tubular epithelial cell apoptosis through YAP/NFAT2 in acute kidney injury mice. Ren Fail 2023; 45:2172961. [PMID: 36718671 PMCID: PMC9891164 DOI: 10.1080/0886022x.2023.2172961] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 02/01/2023] Open
Abstract
AIM Renal tubular epithelial cell (RTEC) apoptosis is important in acute kidney injury (AKI). Calcium/calmodulin-dependent protein kinase II (CaMKII) plays an important role in cell apoptosis, but its potential role in AKI remains unknown. METHODS Using co-immunoprecipitation, immunofluorescence, immunohistochemistry, western blotting, flow cytometry, and cell transfection, this study aimed to verify whether CaMKII is involved in RTEC apoptosis and to explore the underlying mechanism. RESULTS We found that CaMKII was involved in RTEC apoptosis. In adriamycin-induced AKI mice, serum creatinine levels, cell apoptosis, CaMKII activity, and nuclear factor of activated T cells 2 (NFAT2) levels increased, whereas nuclear Yes-associated protein (YAP) expression decreased; inhibition of CaMKII activity reversed these changes. Phosphorylated CaMKII could bind to phosphorylated YAP in the cytoplasm and block it from entering the nucleus, thereby failing to inhibit NFAT2-mediated cell apoptosis. Sequestrated phosphorylated YAP in the RTEC cytoplasm was finally degraded by ubiquitination. CONCLUSION CaMKII may regulate RTEC apoptosis through YAP/NFAT2 in AKI mice. CaMKII may be a potent molecular target for AKI treatment.
Collapse
Affiliation(s)
- Zongshun Huang
- Department of Nephrology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yonghua Peng
- Department of Nephrology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guibao Ke
- Department of Nephrology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yun Xiao
- Department of Nephrology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yaqi Chen
- Department of Nephrology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
62
|
Kerberger R, Brunello G, Drescher D, van Rietbergen B, Becker K. Micro finite element analysis of continuously loaded mini-implants - A micro-CT study in the rat tail model. Bone 2023; 177:116912. [PMID: 37739299 DOI: 10.1016/j.bone.2023.116912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/24/2023]
Abstract
Implant migration has been described as a minor displacement of orthodontic mini-implants (OMIs) when subjected to constant forces. Aim of this study was to evaluate the impact of local stresses on implant migration and bone remodelling around constantly loaded OMIs. Two mini-implants were placed in one caudal vertebra of 61 rats, connected by a nickel‑titanium contraction spring, and loaded with different forces (0.0, 0.5, 1.0, 1.5 N). In vivo micro-CT scans were taken immediately and 1, 2 (n = 61), 4, 6 and 8 (n = 31) weeks post-op. Nine volumes of interest (VOIs) around each implant were defined. To analyse stress values, micro-finite element models were created. Bone remodelling was analysed by calculating the bone volume change between scans performed at consecutive time points. Statistical analysis was performed using a linear mixed model and likelihood-ratio-tests, followed by Tuckey post hoc tests when indicated. The highest stresses were observed in the proximal top VOI. In all VOIs, stress values tended to reach their maximum after two weeks and decreased thereafter. Bone remodelling analysis revealed initial bone loss within the first two weeks and bone gain up to week eight, which was noted especially in the highest loading group. The magnitude of local stresses influenced bone remodelling and it can be speculated that the stress related bone resorption favoured implant migration. After a first healing phase with a high degree of bone resorption, net bone gain representing consolidation was observed.
Collapse
Affiliation(s)
- Robert Kerberger
- Department of Orthodontics, University Hospital Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; Department of Orthodontics and Dentofacial Orthopedics, Charité, Charité Centrum CC03, Institute for Dental and Craniofacial Sciences, Aßmannshauser Straße 4-6, 14197 Berlin, Germany.
| | - Giulia Brunello
- Department of Oral Surgery, University Hospital Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; Department of Neurosciences, School of Dentistry, University of Padova, Via Giustiniani 2, 35128 Padova, Italy.
| | - Dieter Drescher
- Department of Orthodontics, University Hospital Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany.
| | - Bert van Rietbergen
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Groene Loper 3, 5612 AE Eindhoven, the Netherlands.
| | - Kathrin Becker
- Department of Orthodontics, University Hospital Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; Department of Orthodontics and Dentofacial Orthopedics, Charité, Charité Centrum CC03, Institute for Dental and Craniofacial Sciences, Aßmannshauser Straße 4-6, 14197 Berlin, Germany.
| |
Collapse
|
63
|
Hu M, Zhang Y, Zhang X, Zhang X, Huang X, Lu Y, Li Y, Brännström M, Sferruzzi-Perri AN, Shao LR, Billig H. Defective Uterine Spiral Artery Remodeling and Placental Senescence in a Pregnant Rat Model of Polycystic Ovary Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1916-1935. [PMID: 37689383 DOI: 10.1016/j.ajpath.2023.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/28/2023] [Accepted: 08/23/2023] [Indexed: 09/11/2023]
Abstract
Pregnancy-related problems have been linked to impairments in maternal uterine spiral artery (SpA) remodeling. The mechanisms underlying this association are still unclear. It is also unclear whether hyperandrogenism and insulin resistance, the two common manifestations of polycystic ovary syndrome, affect uterine SpA remodeling. We verified previous work in which exposure to 5-dihydrotestosterone (DHT) and insulin (INS) in rats during pregnancy resulted in hyperandrogenism, insulin intolerance, and higher fetal mortality. Exposure to DHT and INS dysregulated the expression of angiogenesis-related genes in the uterus and placenta and also decreased expression of endothelial nitric oxide synthase and matrix metallopeptidases 2 and 9, increased fibrotic collagen deposits in the uterus, and reduced expression of marker genes for SpA-associated trophoblast giant cells. These changes were related to a greater proportion of unremodeled uterine SpAs and a smaller proportion of highly remodeled arteries in DHT + INS-exposed rats. Placentas from DHT + INS-exposed rats exhibited decreased basal and labyrinth zone regions, reduced maternal blood spaces, diminished labyrinth vascularity, and an imbalance in the abundance of vascular and smooth muscle proteins. Furthermore, placentas from DHT + INS-exposed rats showed expression of placental insufficiency markers and a significant increase in cell senescence-associated protein levels. Altogether, this work demonstrates that increased pregnancy complications in polycystic ovary syndrome may be mediated by problems with uterine SpA remodeling, placental functionality, and placental senescence.
Collapse
Affiliation(s)
- Min Hu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China; Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yuehui Zhang
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xu Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - XiuYing Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinyue Huang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yaxing Lu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yijia Li
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Linus R Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Håkan Billig
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
64
|
Cheng X, Barakat R, Pavani G, Usha MK, Calderon R, Snella E, Gorden A, Zhang Y, Gadue P, French DL, Dorman KS, Fidanza A, Campbell CA, Espin-Palazon R. Nod1-dependent NF-kB activation initiates hematopoietic stem cell specification in response to small Rho GTPases. Nat Commun 2023; 14:7668. [PMID: 37996457 PMCID: PMC10667254 DOI: 10.1038/s41467-023-43349-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
Uncovering the mechanisms regulating hematopoietic specification not only would overcome current limitations related to hematopoietic stem and progenitor cell (HSPC) transplantation, but also advance cellular immunotherapies. However, generating functional human induced pluripotent stem cell (hiPSC)-derived HSPCs and their derivatives has been elusive, necessitating a better understanding of the developmental mechanisms that trigger HSPC specification. Here, we reveal that early activation of the Nod1-Ripk2-NF-kB inflammatory pathway in endothelial cells (ECs) primes them to switch fate towards definitive hemogenic endothelium, a pre-requisite to specify HSPCs. Our genetic and chemical embryonic models show that HSPCs fail to specify in the absence of Nod1 and its downstream kinase Ripk2 due to a failure on hemogenic endothelial (HE) programming, and that small Rho GTPases coordinate the activation of this pathway. Manipulation of NOD1 in a human system of definitive hematopoietic differentiation indicates functional conservation. This work establishes the RAC1-NOD1-RIPK2-NF-kB axis as a critical intrinsic inductor that primes ECs prior to HE fate switch and HSPC specification. Manipulation of this pathway could help derive a competent HE amenable to specify functional patient specific HSPCs and their derivatives for the treatment of blood disorders.
Collapse
Affiliation(s)
- Xiaoyi Cheng
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Radwa Barakat
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
- Department of Toxicology, Faculty of Veterinary Medicine, Benha University, Qalyubia, 13518, Egypt
| | - Giulia Pavani
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Masuma Khatun Usha
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Rodolfo Calderon
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Elizabeth Snella
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Abigail Gorden
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Yudi Zhang
- Department of Statistics, Iowa State University, Ames, IA, 50011, USA
| | - Paul Gadue
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Deborah L French
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Karin S Dorman
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
- Department of Statistics, Iowa State University, Ames, IA, 50011, USA
| | - Antonella Fidanza
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, EH16 4UU, Edinburgh, United Kingdom
| | - Clyde A Campbell
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Raquel Espin-Palazon
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
65
|
Li X, Prudente AS, Prato V, Guo X, Hao H, Jones F, Figoli S, Mullen P, Wang Y, Tonnello R, Lee SH, Shah S, Maffei B, Berta T, Du X, Gamper N. Peripheral gating of pain by glial endozepine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567848. [PMID: 38045227 PMCID: PMC10690183 DOI: 10.1101/2023.11.20.567848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
We report that diazepam binding inhibitor (DBI) is a glial messenger mediating satellite glia-sensory neuron crosstalk in the dorsal root ganglion (DRG). DBI is highly and specifically expressed in satellite glia cells (SGCs) of mice, rat and human, but not in sensory neurons or other DRG-resident cells. Knockdown of DBI results in a robust mechanical hypersensitivity without significant effects on other sensory modalities. In vivo overexpression of DBI in SGCs reduces sensitivity to mechanical stimulation and alleviates mechanical allodynia in neuropathic and inflammatory pain models. We further show that DBI acts as a partial agonist and positive allosteric modulator at the neuronal GABAA receptors, particularly strongly effecting those with a high-affinity benzodiazepine binding site. Such receptors are selectively expressed by a subpopulation of mechanosensitive DRG neurons and these are also more enwrapped with DBI-expressing glia, as compared to other DRG neurons, suggesting a mechanism for specific effect of DBI on mechanosensation. These findings identified a new, peripheral neuron-glia communication mechanism modulating pain signalling, which can be targeted therapeutically.
Collapse
Affiliation(s)
- Xinmeng Li
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Vincenzo Prato
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Xianchuan Guo
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Han Hao
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Frederick Jones
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Sofia Figoli
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Pierce Mullen
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Yujin Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Raquel Tonnello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Shihab Shah
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Benito Maffei
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
66
|
Hernández-Silva G, Vega CC, Barrera D, Hernández-Pando R, García-Uribe PÁ, Chirinos M, Larrea F. Gene Expression Changes in the Ovary Mediate Non-Anovulatory Mechanisms of Contraception with Levonorgestrel. Arch Med Res 2023; 54:102892. [PMID: 37804815 DOI: 10.1016/j.arcmed.2023.102892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/06/2023] [Accepted: 09/20/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Emergency contraception with levonorgestrel (LNG) is a viable option to prevent unintended pregnancies. Although the efficacy of LNG as an anovulatory agent decreases as treatment approaches ovulation, it still provides some contraceptive benefits. AIM To better understand the contraceptive mechanisms of LNG in ovulatory subjects. METHODS We conducted a study on Wistar rats that received a single dose of LNG (0.01 or 0.05 mg/kg) on the morning of proestrus before ovulation and evaluated its effects on ovarian gene expression, ovulation, and implantation. RESULTS Our findings showed changes in the expression of genes involved in follicular development and oocyte quality. Pregnancy rates - as an indicator of ovulation - and embryo implantation were significantly lower than those in the control group. CONCLUSIONS This study suggests that LNG alters regulatory factors in the ovary that are essential for the development of competent fertilizable oocytes, highlighting the non-anovulatory mechanisms by which levonorgestrel may regulate fertility and suggesting that it could be a novel observation that contributes to the understanding of emergency contraception in humans.
Collapse
Affiliation(s)
- Gabriela Hernández-Silva
- Reproductive Biology Department Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Claudia Cecilia Vega
- Reproductive Biology Department Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - David Barrera
- Reproductive Biology Department Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Rogelio Hernández-Pando
- Experimental Pathology Lab, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Pablo Ángel García-Uribe
- Reproductive Biology Department Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Mayel Chirinos
- Reproductive Biology Department Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Fernando Larrea
- Reproductive Biology Department Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
67
|
Clark RM, Clark CM, Lewis KE, Dyer MS, Chuckowree JA, Hoyle JA, Blizzard CA, Dickson TC. Intranasal neuropeptide Y1 receptor antagonism improves motor deficits in symptomatic SOD1 ALS mice. Ann Clin Transl Neurol 2023; 10:1985-1999. [PMID: 37644692 PMCID: PMC10647012 DOI: 10.1002/acn3.51885] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
OBJECTIVE Neuropeptide Y (NPY) is a 36 amino acid peptide widely considered to provide neuroprotection in a range of neurodegenerative diseases. In the fatal motor neuron disease amyotrophic lateral sclerosis (ALS), recent evidence supports a link between NPY and ALS disease processes. The goal of this study was to determine the therapeutic potential and role of NPY in ALS, harnessing the brain-targeted intranasal delivery of the peptide, previously utilised to correct motor and cognitive phenotypes in other neurological conditions. METHODS To confirm the association with clinical disease characteristics, NPY expression was quantified in post-mortem motor cortex tissue of ALS patients and age-matched controls. The effect of NPY on ALS cortical pathophysiology was investigated using slice electrophysiology and multi-electrode array recordings of SOD1G93A cortical cultures in vitro. The impact of NPY on ALS disease trajectory was investigated by treating SOD1G93A mice intranasally with NPY and selective NPY receptor agonists and antagonists from pre-symptomatic and symptomatic phases of disease. RESULTS In the human post-mortem ALS motor cortex, we observe a significant increase in NPY expression, which is not present in the somatosensory cortex. In vitro, we demonstrate that NPY can ameliorate ALS hyperexcitability, while brain-targeted nasal delivery of NPY and a selective NPY Y1 receptor antagonist modified survival and motor deficits specifically within the symptomatic phase of the disease in the ALS SOD1G93A mouse. INTERPRETATION Taken together, these findings highlight the capacity for non-invasive brain-targeted interventions in ALS and support antagonism of NPY Y1Rs as a novel strategy to improve ALS motor function.
Collapse
Affiliation(s)
- Rosemary M. Clark
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Courtney M. Clark
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Katherine E.A. Lewis
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Marcus S. Dyer
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Jyoti A. Chuckowree
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Joshua A. Hoyle
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Catherine A. Blizzard
- Tasmanian School of Medicine, College of Health and MedicineUniversity of TasmaniaHobartTasmania7000Australia
| | - Tracey C. Dickson
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| |
Collapse
|
68
|
Shuvaev S, Knipe RS, Drummond M, Rotile NJ, Ay I, Weigand-Whittier JP, Ma H, Zhou IY, Roberts JD, Black K, Hariri LP, Ning Y, Caravan P. Optimization of an Allysine-Targeted PET Probe for Quantifying Fibrogenesis in a Mouse Model of Pulmonary Fibrosis. Mol Imaging Biol 2023; 25:944-953. [PMID: 37610609 DOI: 10.1007/s11307-023-01845-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/14/2023] [Accepted: 08/04/2023] [Indexed: 08/24/2023]
Abstract
PURPOSE Idiopathic pulmonary fibrosis (IPF) is a destructive lung disease with a poor prognosis, an unpredictable clinical course, and inadequate therapies. There are currently no measures of disease activity to guide clinicians making treatment decisions. The aim of this study was to develop a PET probe to identify lung fibrogenesis using a pre-clinical model of pulmonary fibrosis, with potential for translation into clinical use to predict disease progression and inform treatment decisions. METHODS Eight novel allysine-targeting chelators, PIF-1, PIF-2, …, PIF-8, with different aldehyde-reactive moieties were designed, synthesized, and radiolabeled with gallium-68 or copper-64. PET probe performance was assessed in C57BL/6J male mice 2 weeks after intratracheal bleomycin challenge and in naïve mice by dynamic PET/MR imaging and with biodistribution at 90 min post injection. Lung hydroxyproline and allysine were quantified ex vivo and histological staining for fibrosis and aldehyde was performed. RESULTS In vivo screening of probes identified 68GaPIF-3 and 68GaPIF-7 as probes with high uptake in injured lung, high uptake in injured lung versus normal lung, and high uptake in injured lung versus adjacent liver and heart tissue. A crossover, intra-animal PET/MR imaging study of 68GaPIF-3 and 68GaPIF-7 confirmed 68GaPIF-7 as the superior probe. Specificity for fibrogenesis was confirmed in a crossover, intra-animal PET/MR imaging study with 68GaPIF-7 and a non-binding control compound, 68GaPIF-Ctrl. Substituting copper-64 for gallium-68 did not affect lung uptake or specificity indicating that either isotope could be used. CONCLUSION A series of allysine-reactive PET probes with variations in the aldehyde-reactive moiety were evaluated in a pre-clinical model of lung fibrosis. The hydrazine-bearing probe, 68GaPIF-7, exhibited the highest uptake in fibrogenic lung, low uptake in surrounding liver or heart tissue, and low lung uptake in healthy mice and should be considered for further clinical translation.
Collapse
Affiliation(s)
- Sergey Shuvaev
- Institute for Innovation in Imaging (i3), Boston, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Boston, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, Boston, MA, 02129, USA
| | - Rachel S Knipe
- Division of Pulmonary and Critical Care Medicine, Boston, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matt Drummond
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, Boston, MA, 02129, USA
- Division of Pulmonary and Critical Care Medicine, Boston, USA
| | - Nicholas J Rotile
- Institute for Innovation in Imaging (i3), Boston, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Boston, USA
| | - Ilknur Ay
- Institute for Innovation in Imaging (i3), Boston, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Boston, USA
| | | | - Hua Ma
- Institute for Innovation in Imaging (i3), Boston, USA
| | - Iris Yuwen Zhou
- Institute for Innovation in Imaging (i3), Boston, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Boston, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, Boston, MA, 02129, USA
| | - Jesse D Roberts
- Department of Pediatric Anesthesiology, Mass General Hospital for Children, Boston, USA
| | - Katherine Black
- Division of Pulmonary and Critical Care Medicine, Boston, USA
| | - Lida P Hariri
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Yingying Ning
- Institute for Innovation in Imaging (i3), Boston, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Boston, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, Boston, MA, 02129, USA
| | - Peter Caravan
- Institute for Innovation in Imaging (i3), Boston, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Boston, USA.
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, Boston, MA, 02129, USA.
| |
Collapse
|
69
|
Ahn SJ, Anfray A, Anrather J, Iadecola C. Calcium transients in nNOS neurons underlie distinct phases of the neurovascular response to barrel cortex activation in awake mice. J Cereb Blood Flow Metab 2023; 43:1633-1647. [PMID: 37149758 PMCID: PMC10581240 DOI: 10.1177/0271678x231173175] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/14/2023] [Accepted: 04/02/2023] [Indexed: 05/08/2023]
Abstract
Neuronal nitric oxide (NO) synthase (nNOS), a Ca2+ dependent enzyme, is expressed by distinct populations of neocortical neurons. Although neuronal NO is well known to contribute to the blood flow increase evoked by neural activity, the relationships between nNOS neurons activity and vascular responses in the awake state remain unclear. We imaged the barrel cortex in awake, head-fixed mice through a chronically implanted cranial window. The Ca2+ indicator GCaMP7f was expressed selectively in nNOS neurons using adenoviral gene transfer in nNOScre mice. Air-puffs directed at the contralateral whiskers or spontaneous motion induced Ca2+ transients in 30.2 ± 2.2% or 51.6 ± 3.3% of nNOS neurons, respectively, and evoked local arteriolar dilation. The greatest dilatation (14.8 ± 1.1%) occurred when whisking and motion occurred simultaneously. Ca2+ transients in individual nNOS neurons and local arteriolar dilation showed various degrees of correlation, which was strongest when the activity of whole nNOS neuron ensemble was examined. We also found that some nNOS neurons became active immediately prior to arteriolar dilation, while others were activated gradually after arteriolar dilatation. Discrete nNOS neuron subsets may contribute either to the initiation or to the maintenance of the vascular response, suggesting a previously unappreciated temporal specificity to the role of NO in neurovascular coupling.
Collapse
Affiliation(s)
- Sung Ji Ahn
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Antoine Anfray
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
70
|
Umbarkar P, Ejantkar S, Ruiz Ramirez SY, Toro Cora A, Zhang Q, Tousif S, Lal H. Cardiac fibroblast GSK-3α aggravates ischemic cardiac injury by promoting fibrosis, inflammation, and impairing angiogenesis. Basic Res Cardiol 2023; 118:35. [PMID: 37656238 PMCID: PMC11340261 DOI: 10.1007/s00395-023-01005-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023]
Abstract
Myocardial infarction (MI) is the leading cause of death worldwide. Glycogen synthase kinase-3 (GSK-3) has been considered to be a promising therapeutic target for cardiovascular diseases. GSK-3 is a family of ubiquitously expressed serine/threonine kinases. GSK-3 isoforms appear to play overlapping, unique, and even opposing functions in the heart. Previously, our group identified that cardiac fibroblast (FB) GSK-3β acts as a negative regulator of fibrotic remodeling in the ischemic heart. However, the role of FB-GSK-3α in MI pathology is not defined. To determine the role of FB-GSK-3α in MI-induced adverse cardiac remodeling, GSK-3α was deleted specifically in the residential fibroblast or myofibroblast (MyoFB) using tamoxifen (TAM) inducible Tcf21 or Periostin (Postn) promoter-driven Cre recombinase, respectively. Echocardiographic analysis revealed that FB- or MyoFB-specific GSK-3α deletion prevented the development of dilative remodeling and cardiac dysfunction. Morphometrics and histology studies confirmed improvement in capillary density and a remarkable reduction in hypertrophy and fibrosis in the KO group. We harvested the hearts at 4 weeks post-MI and analyzed signature genes of adverse remodeling. Specifically, qPCR analysis was performed to examine the gene panels of inflammation (TNFα, IL-6, IL-1β), fibrosis (COL1A1, COL3A1, COMP, Fibronectin-1, Latent TGF-β binding protein 2), and hypertrophy (ANP, BNP, MYH7). These molecular markers were essentially normalized due to FB-specific GSK-3α deletion. Further molecular studies confirmed that FB-GSK-3α could regulate NF-kB activation and expression of angiogenesis-related proteins. Our findings suggest that FB-GSK-3α plays a critical role in the pathological cardiac remodeling of ischemic hearts, therefore, it could be therapeutically targeted.
Collapse
Affiliation(s)
- Prachi Umbarkar
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA.
| | - Suma Ejantkar
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA
| | - Sulivette Y Ruiz Ramirez
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA
| | - Angelica Toro Cora
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA
| | - Qinkun Zhang
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA
| | - Sultan Tousif
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA
| | - Hind Lal
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA.
| |
Collapse
|
71
|
Li K, Gonye EC, Stornetta RL, Bayliss CB, Yi G, Stornetta DS, Baca SM, Abbott SB, Guyenet PG, Bayliss DA. The astrocytic Na + -HCO 3 - cotransporter, NBCe1, is dispensable for respiratory chemosensitivity. J Physiol 2023; 601:3667-3686. [PMID: 37384821 PMCID: PMC10528273 DOI: 10.1113/jp284960] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
The interoceptive homeostatic mechanism that controls breathing, blood gases and acid-base balance in response to changes in CO2 /H+ is exquisitely sensitive, with convergent roles proposed for chemosensory brainstem neurons in the retrotrapezoid nucleus (RTN) and their supporting glial cells. For astrocytes, a central role for NBCe1, a Na+ -HCO3 - cotransporter encoded by Slc4a4, has been envisaged in multiple mechanistic models (i.e. underlying enhanced CO2 -induced local extracellular acidification or purinergic signalling). We tested these NBCe1-centric models by using conditional knockout mice in which Slc4a4 was deleted from astrocytes. In GFAP-Cre;Slc4a4fl/fl mice we found diminished expression of Slc4a4 in RTN astrocytes by comparison to control littermates, and a concomitant reduction in NBCe1-mediated current. Despite disrupted NBCe1 function in RTN-adjacent astrocytes from these conditional knockout mice, CO2 -induced activation of RTN neurons or astrocytes in vitro and in vivo, and CO2 -stimulated breathing, were indistinguishable from NBCe1-intact littermates; hypoxia-stimulated breathing and sighs were likewise unaffected. We obtained a more widespread deletion of NBCe1 in brainstem astrocytes by using tamoxifen-treated Aldh1l1-Cre/ERT2;Slc4a4fl/fl mice. Again, there was no difference in effects of CO2 or hypoxia on breathing or on neuron/astrocyte activation in NBCe1-deleted mice. These data indicate that astrocytic NBCe1 is not required for the respiratory responses to these chemoreceptor stimuli in mice, and that any physiologically relevant astrocytic contributions must involve NBCe1-independent mechanisms. KEY POINTS: The electrogenic NBCe1 transporter is proposed to mediate local astrocytic CO2 /H+ sensing that enables excitatory modulation of nearby retrotrapezoid nucleus (RTN) neurons to support chemosensory control of breathing. We used two different Cre mouse lines for cell-specific and/or temporally regulated deletion of the NBCe1 gene (Slc4a4) in astrocytes to test this hypothesis. In both mouse lines, Slc4a4 was depleted from RTN-associated astrocytes but CO2 -induced Fos expression (i.e. cell activation) in RTN neurons and local astrocytes was intact. Likewise, respiratory chemoreflexes evoked by changes in CO2 or O2 were unaffected by loss of astrocytic Slc4a4. These data do not support the previously proposed role for NBCe1 in respiratory chemosensitivity mediated by astrocytes.
Collapse
Affiliation(s)
- Keyong Li
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Elizabeth C. Gonye
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Ruth L. Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | | | - Grace Yi
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Daniel S. Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Serapio M. Baca
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Stephen B.G. Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Patrice G. Guyenet
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| | - Douglas A. Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA, 22908
| |
Collapse
|
72
|
Yao Z, Liu N, Lin H, Zhou Y. The Role of miR-1183: A Potential Suppressor in Hepatocellular Carcinoma via Regulating Splicing Factor SRSF1. J Hepatocell Carcinoma 2023; 10:1169-1180. [PMID: 37497429 PMCID: PMC10368139 DOI: 10.2147/jhc.s408542] [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: 02/14/2023] [Accepted: 07/07/2023] [Indexed: 07/28/2023] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) is a severe global health problem, causing many deaths of patients all over the world. Serine and arginine-rich splicing factor 1 (SRSF1) functions as an important oncogenic role in tumorigenesis and progression in HCC. Therefore, therapies targeting SRSF1 may provide promising therapeutic approaches. MiRNAs are virtually involved at the post-transcriptional level and bind to 3' untranslated region (3'-UTR) of their target messenger RNA (mRNA) to suppress expression. Methods Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was used to detect the expression of SRSF1 and miR-1183 in HCC cell lines. CCK8 assay, colony formation assay and wound healing assay were used to detect the function of miR-1183 in HCC cell lines in vitro. Luciferase reporter assay and Western blot were applied to detect the regulation of particular molecules. Xenograft tumor assay was used to detect the function of miR-1183 in HCC cell lines in vivo. Immunohistochemistry (IHC) was used to detect the expression of SRSF1 in HCC tissues and Xenograft tumors. Results In this study, we identified that miR-1183 was downregulated in HCC cell lines. Functional assays indicated that miR-1183-upregulation cells show weakened proliferation ability and migration ability in vitro and inhibit subcutaneous tumor formation in vivo. With respect to the underlying mechanism, we found that miR-1183 function as a tumor suppressor by specifically binding to SRSF1. Conclusion This study is the first to demonstrate that miR-1183 function as an important tumor-suppressing role by binding to the 3'-UTR of SRSF1 mRNA and suppressing its protein level in HCC cells in vitro and in vivo. Further, miR-1183 may be a potential target in the prognosis and treatment of HCC.
Collapse
Affiliation(s)
- Zhilu Yao
- Department of Gastroenterology, Jingan District Zhabei Central Hospital, Shanghai, 200072, People’s Republic of China
- Clinical Medical College of Shanghai Tenth People’s Hospital, Nanjing Medical University, Nanjing, Jiangsu, 211166, People’s Republic of China
| | - Ning Liu
- Department of Gastroenterology, Changzhou Maternal and Child Health Hospital, Changzhou, 213004, People’s Republic of China
| | - Hui Lin
- Department of Gastroenterology, Jingan District Zhabei Central Hospital, Shanghai, 200072, People’s Republic of China
| | - Yingqun Zhou
- Clinical Medical College of Shanghai Tenth People’s Hospital, Nanjing Medical University, Nanjing, Jiangsu, 211166, People’s Republic of China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072, People’s Republic of China
| |
Collapse
|
73
|
Vivona L, Huhle R, Braune A, Scharffenberg M, Wittenstein J, Kiss T, Kircher M, Herzog P, Herzog M, Millone M, Gama de Abreu M, Bluth T. Variable ventilation versus stepwise lung recruitment manoeuvres for lung recruitment: A comparative study in an experimental model of atelectasis. Eur J Anaesthesiol 2023; 40:501-510. [PMID: 36809307 DOI: 10.1097/eja.0000000000001808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
BACKGROUND Variable ventilation recruits alveoli in atelectatic lungs, but it is unknown how it compares with conventional recruitment manoeuvres. OBJECTIVES To test whether mechanical ventilation with variable tidal volumes and conventional recruitment manoeuvres have comparable effects on lung function. DESIGN Randomised crossover study. SETTING University hospital research facility. ANIMALS Eleven juvenile mechanically ventilated pigs with atelectasis created by saline lung lavage. INTERVENTIONS Lung recruitment was performed using two strategies, both with an individualised optimal positive-end expiratory pressure (PEEP) associated with the best respiratory system elastance during a decremental PEEP trial: conventional recruitment manoeuvres (stepwise increase of PEEP) in pressure-controlled mode) followed by 50 min of volume-controlled ventilation (VCV) with constant tidal volume, and variable ventilation, consisting of 50 min of VCV with random variation in tidal volume. MAIN OUTCOME MEASURES Before and 50 min after each recruitment manoeuvre strategy, lung aeration was assessed by computed tomography, and relative lung perfusion and ventilation (0% = dorsal, 100% = ventral) were determined by electrical impedance tomography. RESULTS After 50 min, variable ventilation and stepwise recruitment manoeuvres decreased the relative mass of poorly and nonaerated lung tissue (percent lung mass: 35.3 ± 6.2 versus 34.2 ± 6.6, P = 0.303); reduced poorly aerated lung mass compared with baseline (-3.5 ± 4.0%, P = 0.016, and -5.2 ± 2.8%, P < 0.001, respectively), and reduced nonaerated lung mass compared with baseline (-7.2 ± 2.5%, P < 0.001; and -4.7 ± 2.8%, P < 0.001 respectively), while the distribution of relative perfusion was barely affected (variable ventilation: -0.8 ± 1.1%, P = 0.044; stepwise recruitment manoeuvres: -0.4 ± 0.9%, P = 0.167). Compared with baseline, variable ventilation and stepwise recruitment manoeuvres increased Pa O 2 (172 ± 85mmHg, P = 0.001; and 213 ± 73 mmHg, P < 0.001, respectively), reduced Pa CO 2 (-9.6 ± 8.1 mmHg, P = 0.003; and -6.7 ± 4.6 mmHg, P < 0.001, respectively), and decreased elastance (-11.4 ± 6.3 cmH 2 O, P < 0.001; and -14.1 ± 3.3 cmH 2 O, P < 0.001, respectively). Mean arterial pressure decreased during stepwise recruitment manoeuvres (-24 ± 8 mmHg, P = 0.006), but not variable ventilation. CONCLUSION In this model of lung atelectasis, variable ventilation and stepwise recruitment manoeuvres effectively recruited lungs, but only variable ventilation did not adversely affect haemodynamics. TRIAL REGISTRATION This study was registered and approved by Landesdirektion Dresden, Germany (DD24-5131/354/64).
Collapse
Affiliation(s)
- Luigi Vivona
- From the Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany (LV, RH, AB, MS, JW, TK, PH, MH, MM, MGA, TB), Department of Pathophysiology and Transplantation, University of Milan, Italy (LV), Institute of Nuclear Medicine, University Hospital Carl Gustav Carus, Dresden (AB), Department of Anesthesiology, Elblandklinikum Radebeul, Radebeul (TK), Institute of Biomedical Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe (MK), Drägerwerk AG & Co KGaA, Lübeck, Germany (MK), IRCCS San Martino IST, Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy (MM), Department of Intensive Care and Resuscitation (MGA) and Department of Outcomes Research, Anesthesiology Institute, Cleveland Clinic, Cleveland, Ohio, USA (MGA)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Gugliandolo E, Macrì F, Fusco R, Siracusa R, Cordaro M, D'amico R, Peritore AF, Impellizzeri D, Genovese T, Cuzzocrea S, Di Paola R, Crupi R. Inhibiting IL-6 in medicine: a new twist to sustain inhibition of his cytokine tin the therapy of Pulmonary Arterial Hypertension. Pharmacol Res 2023; 192:106750. [PMID: 37004831 DOI: 10.1016/j.phrs.2023.106750] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/18/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a chronic, progressive disease characterized by an increase in blood pressure in the lungs' arteries. It can occur in a variety of species, including humans, dogs, cats, and horses. To date, PAH has a high mortality rate in both veterinary and human medicine, often due to complications such as heart failure. The complex pathological mechanisms of PAH involve multiple cellular signalling pathways at various levels. IL-6 is a powerful pleiotropic cytokine that regulates several phases of immune response, inflammation, and tissue remodelling. The hypothesis of this study was that the use of an IL-6 antagonist in PAH could interrupt or mitigate the cascade of events that leads to the progression of the disease and the worsening of clinical outcome, as well as tissue remodelling. In this study, we used two pharmacological protocols with an IL-6 receptor antagonist in a monocrotaline-induced PAH model in rats. Our results showed that the use of an IL-6 receptor antagonist had a significant protective effect, ameliorating both haemodynamic parameters, lung and cardiac function, tissue remodelling, and the inflammation associated with PAH. The results of this study suggest that the inhibition IL-6 could be a useful pharmacological strategy in PAH, in both human and veterinary medicine.
Collapse
Affiliation(s)
- Enrico Gugliandolo
- Department of Veterinary Science, University of Messina, Via Giovanni Palatucci, 98168 Messina, Italy.
| | - Francesco Macrì
- Department of Veterinary Science, University of Messina, Via Giovanni Palatucci, 98168 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98166 Messina, Italy
| | - Ramona D'amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy.
| | - Rosanna Di Paola
- Department of Veterinary Science, University of Messina, Via Giovanni Palatucci, 98168 Messina, Italy
| | - Rosalia Crupi
- Department of Veterinary Science, University of Messina, Via Giovanni Palatucci, 98168 Messina, Italy
| |
Collapse
|
75
|
White A, Stremming J, Brown LD, Rozance PJ. Attenuated glucose-stimulated insulin secretion during an acute IGF-1 LR3 infusion into fetal sheep does not persist in isolated islets. J Dev Orig Health Dis 2023; 14:353-361. [PMID: 37114757 PMCID: PMC10205682 DOI: 10.1017/s2040174423000090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Insulin-like growth factor-1 (IGF-1) is a critical fetal growth hormone that has been proposed as a therapy for intrauterine growth restriction. We previously demonstrated that a 1-week IGF-1 LR3 infusion into fetal sheep reduces in vivo and in vitro insulin secretion suggesting an intrinsic islet defect. Our objective herein was to determine whether this intrinsic islet defect was related to chronicity of exposure. We therefore tested the effects of a 90-min IGF-1 LR3 infusion on fetal glucose-stimulated insulin secretion (GSIS) and insulin secretion from isolated fetal islets. We first infused late gestation fetal sheep (n = 10) with either IGF-1 LR3 (IGF-1) or vehicle control (CON) and measured basal insulin secretion and in vivo GSIS utilizing a hyperglycemic clamp. We then isolated fetal islets immediately following a 90-min IGF-1 or CON in vivo infusion and exposed them to glucose or potassium chloride to measure in vitro insulin secretion (IGF-1, n = 6; CON, n = 6). Fetal plasma insulin concentrations decreased with IGF-1 LR3 infusion (P < 0.05), and insulin concentrations during the hyperglycemic clamp were 66% lower with IGF-1 LR3 infusion compared to CON (P < 0.0001). Insulin secretion in isolated fetal islets was not different based on infusion at the time of islet collection. Therefore, we speculate that while acute IGF-1 LR3 infusion may directly suppress insulin secretion, the fetal β-cell in vitro retains the ability to recover GSIS. This may have important implications when considering the long-term effects of treatment modalities for fetal growth restriction.
Collapse
Affiliation(s)
- Alicia White
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jane Stremming
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Laura D Brown
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paul J Rozance
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
76
|
Yu H, Wang M, Yang Q, Xu X, Zhang R, Chen X, Le W. The electrophysiological and neuropathological profiles of cerebellum in APP swe /PS1 ΔE9 mice: A hypothesis on the role of cerebellum in Alzheimer's disease. Alzheimers Dement 2023; 19:2365-2375. [PMID: 36469008 DOI: 10.1002/alz.12853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/28/2022] [Accepted: 10/05/2022] [Indexed: 12/11/2022]
Abstract
We propose the hypothesis that the cerebellar electrophysiology and sleep-wake cycles may be altered at the early stage of Alzheimer's disease (AD), proceeding the amyloid-β neuropathological hallmarks. The electrophysiologic characteristics of cerebellum thereby might be served as a biomarker in the prepathological detection of AD. Sleep disturbances are common in preclinical AD patients, and the cerebellum has been implicated in sleep-wake regulation by several pioneer studies. Additionally, recent studies suggest that the structure and function of the cerebellum may be altered at the early stages of AD, indicating that the cerebellum may be involved in the disease's progression. We used APPswe /PS1ΔE9 mice as a model of AD, monitored and analyzed electroencephalogram data, and assessed neuropathological profiles in the cerebellum of AD mice. Our hypothesis may establish a linkage between the cerebellum and AD, thereby potentially providing new perspectives on the pathogenesis of the disease.
Collapse
Affiliation(s)
- Hang Yu
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Manli Wang
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Qiu Yang
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Xiaojiao Xu
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Rong Zhang
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Xi Chen
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Weidong Le
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
77
|
Kelly SB, Dean JM, Zahra VA, Dudink I, Thiel A, Polglase GR, Miller SL, Hooper SB, Bennet L, Gunn AJ, Galinsky R. Progressive inflammation reduces high-frequency EEG activity and cortical dendritic arborisation in late gestation fetal sheep. J Neuroinflammation 2023; 20:124. [PMID: 37226206 DOI: 10.1186/s12974-023-02805-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Antenatal infection/inflammation is associated with disturbances in neuronal connectivity, impaired cortical growth and poor neurodevelopmental outcomes. The pathophysiological substrate that underpins these changes is poorly understood. We tested the hypothesis that progressive inflammation in late gestation fetal sheep would alter cortical neuronal microstructure and neural function assessed using electroencephalogram band power analysis. METHODS Fetal sheep (0.85 of gestation) were surgically instrumented for continuous electroencephalogram (EEG) recording and randomly assigned to repeated saline (control; n = 9) or LPS (0 h = 300 ng, 24 h = 600 ng, 48 h = 1200 ng; n = 8) infusions to induce inflammation. Sheep were euthanised 4 days after the first LPS infusion for assessment of inflammatory gene expression, histopathology and neuronal dendritic morphology in the somatosensory cortex. RESULTS LPS infusions increased delta power between 8 and 50 h, with reduced beta power from 18 to 96 h (P < 0.05 vs. control). Basal dendritic length, numbers of dendritic terminals, dendritic arborisation and numbers of dendritic spines were reduced in LPS-exposed fetuses (P < 0.05 vs. control) within the somatosensory cortex. Numbers of microglia and interleukin (IL)-1β immunoreactivity were increased in LPS-exposed fetuses compared with controls (P < 0.05). There were no differences in total numbers of cortical NeuN + neurons or cortical area between the groups. CONCLUSIONS Exposure to antenatal infection/inflammation was associated with impaired dendritic arborisation, spine number and loss of high-frequency EEG activity, despite normal numbers of neurons, that may contribute to disturbed cortical development and connectivity.
Collapse
Affiliation(s)
- Sharmony B Kelly
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Justin M Dean
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Valerie A Zahra
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
| | - Ingrid Dudink
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Alison Thiel
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Laura Bennet
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
78
|
Zhou S, Yu Z, Chen Z, Ning F, Hu X, Wu T, Li M, Xin H, Reilly S, Zhang X. Olmesartan alleviates SARS-CoV-2 envelope protein induced renal fibrosis by regulating HMGB1 release and autophagic degradation of TGF-β1. Front Pharmacol 2023; 14:1187818. [PMID: 37256223 PMCID: PMC10225711 DOI: 10.3389/fphar.2023.1187818] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/09/2023] [Indexed: 06/01/2023] Open
Abstract
Background and aims: Renal damage in severe coronavirus disease 2019 (COVID-19) is highly associated with mortality. Finding relevant therapeutic candidates that can alleviate it is crucial. Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin-receptor blockers (ARBs) have been shown to be harmless to COVID-19 patients, but it remains elusive whether ACEIs/ARBs have protective benefits to them. We wished to determine if ACEIs/ARBs had a protective effect on the renal damage associated with COVID-19, and to investigate the mechanism. Methods: We used the envelope (E) protein of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) to induce COVID-19-like multiple organ damage and observed renal fibrosis. We induced the epithelial-mesenchymal transformation of HK-2 cells with E protein, and found that olmesartan could alleviate it significantly. The protective effects of olmesartan on E protein-induced renal fibrosis were evaluated by renal-function assessment, pathologic alterations, inflammation, and the TGF-β1/Smad2/3 signaling pathway. The distribution of high-mobility group box (HMGB)1 was examined after stimulation with E protein and olmesartan administration. Results: E protein stimulated HMGB1 release, which triggered the immune response and promoted activation of TGF-β1/Smad2/3 signaling: both could lead to renal fibrosis. Olmesartan regulated the distribution of HMGB1 under E protein stimulation. Olmesartan inhibited the release of HMGB1, and reduced the inflammatory response and activation of TGF-β1/Smad2/3 signaling. Olmesartan increased the cytoplasmic level of HMGB1 to promote the autophagic degradation of TGF-β1, thereby alleviating fibrosis further. Conclusion: Olmesartan alleviates E protein-induced renal fibrosis by regulating the release of HMGB1 and its mediated autophagic degradation of TGF-β1.
Collapse
Affiliation(s)
- Shilin Zhou
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Zanzhe Yu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zihui Chen
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Fengling Ning
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xuetao Hu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Tiangang Wu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Mingxue Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
79
|
Tran KKN, Wong VHY, Hoang A, Finkelstein DI, Bui BV, Nguyen CTO. Retinal alpha-synuclein accumulation correlates with retinal dysfunction and structural thinning in the A53T mouse model of Parkinson's disease. Front Neurosci 2023; 17:1146979. [PMID: 37214398 PMCID: PMC10196133 DOI: 10.3389/fnins.2023.1146979] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 04/17/2023] [Indexed: 05/24/2023] Open
Abstract
Abnormal alpha-synuclein (α-SYN) protein deposition has long been recognized as one of the pathological hallmarks of Parkinson's disease's (PD). This study considers the potential utility of PD retinal biomarkers by investigating retinal changes in a well characterized PD model of α-SYN overexpression and how these correspond to the presence of retinal α-SYN. Transgenic A53T homozygous (HOM) mice overexpressing human α-SYN and wildtype (WT) control littermates were assessed at 4, 6, and 14 months of age (male and female, n = 15-29 per group). In vivo retinal function (electroretinography, ERG) and structure (optical coherence tomography, OCT) were recorded, and retinal immunohistochemistry and western blot assays were performed to examine retinal α-SYN and tyrosine hydroxylase. Compared to WT controls, A53T mice exhibited reduced light-adapted (cone photoreceptor and bipolar cell amplitude, p < 0.0001) ERG responses and outer retinal thinning (outer plexiform layer, outer nuclear layer, p < 0.0001) which correlated with elevated levels of α-SYN. These retinal signatures provide a high throughput means to study α-SYN induced neurodegeneration and may be useful in vivo endpoints for PD drug discovery.
Collapse
Affiliation(s)
- Katie K. N. Tran
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Vickie H. Y. Wong
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Anh Hoang
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Bang V. Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Christine T. O. Nguyen
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
80
|
Galinsky R, Dhillon SK, Kelly SB, Wassink G, Davidson JO, Lear CA, van den Heuij LG, Bennet L, Gunn AJ. Magnesium sulphate reduces tertiary gliosis but does not improve EEG recovery or white or grey matter cell survival after asphyxia in preterm fetal sheep. J Physiol 2023; 601:1999-2016. [PMID: 36999348 PMCID: PMC10952359 DOI: 10.1113/jp284381] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/09/2023] [Indexed: 04/01/2023] Open
Abstract
Maternal magnesium sulphate (MgSO4 ) treatment is widely recommended before preterm birth for neuroprotection. However, this is controversial because there is limited evidence that MgSO4 provides long-term neuroprotection. Preterm fetal sheep (104 days gestation; term is 147 days) were assigned randomly to receive sham occlusion with saline infusion (n = 6) or i.v. infusion with MgSO4 (n = 7) or vehicle (saline, n = 6) from 24 h before hypoxia-ischaemia induced by umbilical cord occlusion until 24 h after occlusion. Sheep were killed after 21 days of recovery, for fetal brain histology. Functionally, MgSO4 did not improve long-term EEG recovery. Histologically, in the premotor cortex and striatum, MgSO4 infusion attenuated post-occlusion astrocytosis (GFAP+ ) and microgliosis but did not affect numbers of amoeboid microglia or improve neuronal survival. In the periventricular and intragyral white matter, MgSO4 was associated with fewer total (Olig-2+ ) oligodendrocytes compared with vehicle + occlusion. Numbers of mature (CC1+ ) oligodendrocytes were reduced to a similar extent in both occlusion groups compared with sham occlusion. In contrast, MgSO4 was associated with an intermediate improvement in myelin density in the intragyral and periventricular white matter tracts. In conclusion, a clinically comparable dose of MgSO4 was associated with moderate improvements in white and grey matter gliosis and myelin density but did not improve EEG maturation or neuronal or oligodendrocyte survival. KEY POINTS: Magnesium sulphate is widely recommended before preterm birth for neuroprotection; however, there is limited evidence that magnesium sulphate provides long-term neuroprotection. In preterm fetal sheep exposed to hypoxia-ischaemia (HI), MgSO4 was associated with attenuated astrocytosis and microgliosis in the premotor cortex and striatum but did not improve neuronal survival after recovery to term-equivalent age, 21 days after HI. Magnesium sulphate was associated with loss of total oligodendrocytes in the periventricular and intragyral white matter tracts, whereas mature, myelinating oligodendrocytes were reduced to a similar extent in both occlusion groups. In the same regions, MgSO4 was associated with an intermediate improvement in myelin density. Functionally, MgSO4 did not improve long-term recovery of EEG power, frequency or sleep stage cycling. A clinically comparable dose of MgSO4 was associated with moderate improvements in white and grey matter gliosis and myelin density but did not improve EEG maturation or neuronal or oligodendrocyte survival.
Collapse
Affiliation(s)
- Robert Galinsky
- Department of PhysiologyUniversity of AucklandAucklandNew Zealand
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Obstetrics and GynaecologyMonash UniversityVictoriaAustralia
| | | | - Sharmony B. Kelly
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Obstetrics and GynaecologyMonash UniversityVictoriaAustralia
| | - Guido Wassink
- Department of PhysiologyUniversity of AucklandAucklandNew Zealand
| | | | | | | | - Laura Bennet
- Department of PhysiologyUniversity of AucklandAucklandNew Zealand
| | - Alistair J. Gunn
- Department of PhysiologyUniversity of AucklandAucklandNew Zealand
| |
Collapse
|
81
|
Halade GV, Mat Y, Gowda SGB, Jain S, Hui S, Yadav H, Kain V. Sleep deprivation in obesogenic setting alters lipidome and microbiome toward suboptimal inflammation in acute heart failure. FASEB J 2023; 37:e22899. [PMID: 37002889 DOI: 10.1096/fj.202300184r] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/10/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
Sleep is a fundamental medicine for cardiac homeostasis, and sleep-deprived individuals are prone to higher incidences of heart attack. The lipid-dense diet (obesogenic diet-OBD) is a cumulative risk factor for chronic inflammation in cardiovascular disease; thus, understanding how sleep fragmentation (SF) in an obesity setting impacts immune and cardiac health is an unmet medical need. We hypothesized whether the co-existence of SF with OBD dysregulates gut homeostasis and leukocyte-derived reparative/resolution mediators, thereby impairing cardiac repair. Two-month-old male C57BL/6J mice were randomized first into two groups, then four groups; Control, control + SF, OBD, and OBD + SF mice subjected to myocardial infarction (MI). OBD mice had higher levels of plasma linolenic acid with a decrease in eicosapentaenoic and docosahexaenoic acid. The OBD mice had lower Lactobacillus johnsonii indicating a loss of probiotic microbiota. SF in OBD mice increased Firmicutes/Bacteroidetes ratio indicative of a detrimental change in SF-directed microbiome. OBD + SF group increased in the neutrophil: lymphocyte ratio suggestive of suboptimal inflammation. As a result of SF, resolution mediators (RvD2, RvD3, RvD5, LXA4 , PD1, and MaR1) decreased and inflammatory mediators (PGD2 , PGE2 , PGF2a , 6k-PGF1a ) were increased in OBD mice post-MI. At the site of infarction, the proinflammatory cytokines Ccl2, IL1β, and IL-6 were amplified in OBD + SF indicating a robust proinflammatory milieu post-MI. Also, brain circadian genes (Bmal1, Clock) were downregulated in SF-subjected control mice, but remained elevated in OBD mice post-MI. SF superimposed on obesity dysregulated physiological inflammation and disrupted resolving response thereby impaired cardiac repair and signs of pathological inflammation.
Collapse
Affiliation(s)
- Ganesh V. Halade
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine University of South Florida Tampa Florida USA
| | - Yusuf Mat
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine University of South Florida Tampa Florida USA
| | | | - Shalini Jain
- USF Center for Microbiome Research Microbiomes Institute Tampa Florida USA
- Center for Aging and Brain Repair University of South Florida Tampa Florida USA
| | - Shu‐Ping Hui
- Faculty of Health Sciences Hokkaido University Sapporo Japan
| | - Hariom Yadav
- USF Center for Microbiome Research Microbiomes Institute Tampa Florida USA
- Center for Aging and Brain Repair University of South Florida Tampa Florida USA
| | - Vasundhara Kain
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine University of South Florida Tampa Florida USA
| |
Collapse
|
82
|
Kirk ME, Merit VT, Moeslund N, Dragsbaek SJ, Hansen JV, Andersen A, Lyhne MD. Impact of sternotomy and pericardiotomy on cardiopulmonary haemodynamics in a large animal model. Exp Physiol 2023; 108:762-771. [PMID: 36892095 PMCID: PMC10988510 DOI: 10.1113/ep090919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/14/2023] [Indexed: 03/10/2023]
Abstract
NEW FINDINGS What is the central question of this study? Invasive cardiovascular instrumentation can occur through closed- or open-chest approaches. To what extent will sternotomy and pericardiotomy affect cardiopulmonary variables? What is the main finding and its importance? Opening of the thorax decreased mean systemic and pulmonary pressures. Left ventricular function improved, but no changes were observed in right ventricular systolic measures. No consensus or recommendation exists regarding instrumentation. Methodological differences risk compromising rigour and reproducibility in preclinical research. ABSTRACT Animal models of cardiovascular disease are often evaluated by invasive instrumentation for phenotyping. As no consensus exists, both open- and closed-chest approaches are used, which might compromise rigour and reproducibility in preclinical research. We aimed to quantify the cardiopulmonary changes induced by sternotomy and pericardiotomy in a large animal model. Seven pigs were anaesthetized, mechanically ventilated and evaluated by right heart catheterization and bi-ventricular pressure-volume loop recordings at baseline and after sternotomy and pericardiotomy. Data were compared by ANOVA or the Friedmann test where appropriate, with post-hoc analyses to control for multiple comparisons. Sternotomy and pericardiotomy caused reductions in mean systemic (-12 ± 11 mmHg, P = 0.027) and pulmonary pressures (-4 ± 3 mmHg, P = 0.006) and airway pressures. Cardiac output decreased non-significantly (-1329 ± 1762 ml/min, P = 0.052). Left ventricular afterload decreased, with an increase in ejection fraction (+9 ± 7%, P = 0.027) and coupling. No changes were observed in right ventricular systolic function or arterial blood gases. In conclusion, open- versus closed-chest approaches to invasive cardiovascular phenotyping cause a systematic difference in key haemodynamic variables. Researchers should adopt the most appropriate approach to ensure rigour and reproducibility in preclinical cardiovascular research.
Collapse
Affiliation(s)
- Mathilde Emilie Kirk
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of CardiologyAarhus University HospitalAarhusDenmark
| | - Victor Tang Merit
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of CardiologyAarhus University HospitalAarhusDenmark
| | - Niels Moeslund
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Cardiac, Lung and Vascular SurgeryAarhus University HospitalAarhusDenmark
| | - Simone Juel Dragsbaek
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of CardiologyAarhus University HospitalAarhusDenmark
| | - Jacob Valentin Hansen
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of CardiologyAarhus University HospitalAarhusDenmark
| | - Asger Andersen
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of CardiologyAarhus University HospitalAarhusDenmark
| | - Mads Dam Lyhne
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Anaesthesiology and Intensive CareAarhus University HospitalAarhusDenmark
| |
Collapse
|
83
|
Lear CA, Beacom MJ, Dhillon SK, Lear BA, Mills OJ, Gunning MI, Westgate JA, Bennet L, Gunn AJ. Dissecting the contributions of the peripheral chemoreflex and myocardial hypoxia to fetal heart rate decelerations in near-term fetal sheep. J Physiol 2023; 601:2017-2041. [PMID: 37017488 DOI: 10.1113/jp284286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/30/2023] [Indexed: 04/06/2023] Open
Abstract
Brief repeated fetal hypoxaemia during labour can trigger intrapartum decelerations of the fetal heart rate (FHR) via the peripheral chemoreflex or the direct effects of myocardial hypoxia, but the relative contribution of these two mechanisms and how this balance changes with evolving fetal compromise remain unknown. In the present study, chronically instrumented near-term fetal sheep received surgical vagotomy (n = 8) or sham vagotomy (control, n = 11) to disable the peripheral chemoreflex and unmask myocardial hypoxia. One-minute complete umbilical cord occlusions (UCOs) were performed every 2.5 min for 4 h or until arterial pressure fell below 20 mmHg. Hypotension and severe acidaemia developed progressively after 65.7 ± 7.2 UCOs in control fetuses and 49.5 ± 7.8 UCOs after vagotomy. Vagotomy was associated with faster development of metabolic acidaemia and faster impairment of arterial pressure during UCOs without impairing centralization of blood flow or neurophysiological adaptation to UCOs. During the first half of the UCO series, before severe hypotension developed, vagotomy was associated with a marked increase in FHR during UCOs. After the onset of evolving severe hypotension, FHR fell faster in control fetuses during the first 20 s of UCOs, but FHR during the final 40 s of UCOs became progressively more similar between groups, with no difference in the nadir of decelerations. In conclusion, FHR decelerations were initiated and sustained by the peripheral chemoreflex at a time when fetuses were able to maintain arterial pressure. After the onset of evolving hypotension and acidaemia, the peripheral chemoreflex continued to initiate decelerations, but myocardial hypoxia became progressively more important in sustaining and deepening decelerations. KEY POINTS: Brief repeated hypoxaemia during labour can trigger fetal heart rate decelerations by either the peripheral chemoreflex or myocardial hypoxia, but how this balance changes with fetal compromise is unknown. Reflex control of fetal heart rate was disabled by vagotomy to unmask the effects of myocardial hypoxia in chronically instrumented fetal sheep. Fetuses were then subjected to repeated brief hypoxaemia consistent with the rates of uterine contractions during labour. We show that the peripheral chemoreflex controls brief decelerations in their entirety at a time when fetuses were able to maintain normal or increased arterial pressure. The peripheral chemoreflex still initiated decelerations even after the onset of evolving hypotension and acidaemia, but myocardial hypoxia made an increasing contribution to sustain and deepen decelerations.
Collapse
Affiliation(s)
- Christopher A Lear
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Michael J Beacom
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Simerdeep K Dhillon
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Benjamin A Lear
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Olivia J Mills
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Mark I Gunning
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Jenny A Westgate
- Department of Obstetrics and Gynaecology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
- Department of Paediatrics, Starship Children's Hospital, Auckland, New Zealand
| |
Collapse
|
84
|
Vidaurre MDPH, Osborn BK, Lowak KD, McDonald MM, Wang YWW, Pa V, Richter JR, Xu Y, Arnold K, Liu J, Cardenas JC. A 3- O-sulfated heparan sulfate dodecasaccharide (12-mer) suppresses thromboinflammation and attenuates early organ injury following trauma and hemorrhagic shock. Front Immunol 2023; 14:1158457. [PMID: 37122735 PMCID: PMC10140401 DOI: 10.3389/fimmu.2023.1158457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Dysregulated inflammation and coagulation are underlying mechanisms driving organ injury after trauma and hemorrhagic shock. Heparan sulfates, cell surface glycosaminoglycans abundantly expressed on the endothelial surface, regulate a variety of cellular processes. Endothelial heparan sulfate containing a rare 3-O-sulfate modification on a glucosamine residue is anticoagulant and anti-inflammatory through high-affinity antithrombin binding and sequestering of circulating damage-associated molecular pattern molecules. Our goal was to evaluate therapeutic potential of a synthetic 3-O-sulfated heparan sulfate dodecasaccharide (12-mer, or dekaparin) to attenuate thromboinflammation and prevent organ injury. Methods Male Sprague-Dawley rats were pre-treated subcutaneously with vehicle (saline) or dekaparin (2 mg/kg) and subjected to a trauma/hemorrhagic shock model through laparotomy, gut distention, and fixed-pressure hemorrhage. Vehicle and dekaparin-treated rats were resuscitated with Lactated Ringer's solution (LR) and compared to vehicle-treated fresh-frozen-plasma-(FFP)-resuscitated rats. Serial blood samples were collected at baseline, after induction of shock, and 3 hours after fluid resuscitation to measure hemodynamic and metabolic shock indicators, inflammatory mediators, and thrombin-antithrombin complex formation. Lungs and kidneys were processed for organ injury scoring and immunohistochemical analysis to quantify presence of neutrophils. Results Induction of trauma and hemorrhagic shock resulted in significant increases in thrombin-antithrombin complex, inflammatory markers, and lung and kidney injury scores. Compared to vehicle, dekaparin treatment did not affect induction, severity, or recovery of shock as indicated by hemodynamics, metabolic indicators of shock (lactate and base excess), or metrics of bleeding, including overall blood loss, resuscitation volume, or hematocrit. While LR-vehicle-resuscitated rodents exhibited increased lung and kidney injury, administration of dekaparin significantly reduced organ injury scores and was similar to organ protection conferred by FFP resuscitation. This was associated with a significant reduction in neutrophil infiltration in lungs and kidneys and reduced lung fibrin deposition among dekaparin-treated rats compared to vehicle. No differences in organ injury, neutrophil infiltrates, or fibrin staining between dekaparin and FFP groups were observed. Finally, dekaparin treatment attenuated induction of thrombin-antithrombin complex and inflammatory mediators in plasma following trauma and hemorrhagic shock. Conclusion Anti-thromboinflammatory properties of a synthetic 3-O-sulfated heparan sulfate 12-mer, dekaparin, could provide therapeutic benefit for mitigating organ injury following major trauma and hemorrhagic shock.
Collapse
Affiliation(s)
- Maria del Pilar Huby Vidaurre
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Baron K. Osborn
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Kaylie D. Lowak
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Michelle M. McDonald
- Department of Pathology and Laboratory Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yao-Wei W. Wang
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Veda Pa
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jillian R. Richter
- Department of Surgery, Division of Trauma and Acute Care Surgery, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yongmei Xu
- Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Katelyn Arnold
- Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jian Liu
- Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jessica C. Cardenas
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
85
|
Vammen L, Johannsen CM, Baltsen CD, Nørholt C, Eggertsen M, Mortensen S, Vormfenne L, Povlsen A, Donnino MW, Løfgren B, Andersen LW, Granfeldt A. Thiamine for the Treatment of Cardiac Arrest-Induced Neurological Injury: A Randomized, Blinded, Placebo-Controlled Experimental Study. J Am Heart Assoc 2023; 12:e028558. [PMID: 36942758 PMCID: PMC10122898 DOI: 10.1161/jaha.122.028558] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/06/2023] [Indexed: 03/23/2023]
Abstract
Background Thiamine supplementation has demonstrated protective effects in a mouse model of cardiac arrest. The aim of this study was to investigate the neuroprotective effects of thiamine in a clinically relevant large animal cardiac arrest model. The hypothesis was that thiamine reduces neurological injury evaluated by neuron-specific enolase levels. Methods and Results Pigs underwent myocardial infarction and subsequently 9 minutes of untreated cardiac arrest. Twenty minutes after successful resuscitation, the pigs were randomized to treatment with either thiamine or placebo. All pigs underwent 40 hours of intensive care and were awakened for assessment of functional neurological outcome up until 9 days after cardiac arrest. Nine pigs were included in both groups, with 8 in each group surviving the entire intensive care phase. Mean area under the curve for neuron-specific enolase was similar between groups, with 81.5 μg/L per hour (SD, 20.4) in the thiamine group and 80.5 μg/L per hour (SD, 18.3) in the placebo group, with an absolute difference of 1.0 (95% CI, -57.8 to 59.8; P=0.97). Likewise, there were no absolute difference in neurological deficit score at the end of the protocol (2 [95% CI, -38 to 42]; P=0.93). There was no absolute mean group difference in lactate during the intensive care period (1.1 mmol/L [95% CI, -0.5 to 2.7]; P=0.16). Conclusions In this randomized, blinded, placebo-controlled trial using a pig cardiac arrest model with myocardial infarction and long intensive care and observation for 9 days, thiamine showed no effect in changes to functional neurological outcome or serum levels of neuron-specific enolase. Thiamine treatment had no effect on lactate levels after successful resuscitation.
Collapse
Affiliation(s)
- Lauge Vammen
- Department of Anesthesiology and Intensive CareAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Cecilie Munch Johannsen
- Department of Anesthesiology and Intensive CareAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | | | - Casper Nørholt
- Department of Anesthesiology and Intensive CareAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Mark Eggertsen
- Department of Anesthesiology and Intensive CareAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Signe Mortensen
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Lasse Vormfenne
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Amalie Povlsen
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Cardiothoracic AnesthesiaCopenhagen University Hospital, RigshospitaletRisskovDenmark
| | - Michael W. Donnino
- Center for Resuscitation Science, Department of Emergency MedicineBeth Israel Deaconess Medical CenterBostonMAUSA
- Department of Internal Medicine, Division of PulmonaryCritical Care, and Sleep Medicine, Beth Israel Deaconess Medical CenterBostonMAUSA
| | - Bo Løfgren
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Research Center for Emergency MedicineAarhus University HospitalAarhusDenmark
- Department of MedicineRanders Regional HospitalRandersDenmark
| | - Lars W. Andersen
- Department of Anesthesiology and Intensive CareAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Prehospital Emergency Medical ServicesCentral Denmark RegionAarhusDenmark
| | - Asger Granfeldt
- Department of Anesthesiology and Intensive CareAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
86
|
Garcia-Bonilla L, Shahanoor Z, Sciortino R, Nazarzoda O, Racchumi G, Iadecola C, Anrather J. Brain and blood single-cell transcriptomics in acute and subacute phases after experimental stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.31.535150. [PMID: 37066298 PMCID: PMC10103945 DOI: 10.1101/2023.03.31.535150] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Cerebral ischemia triggers a powerful inflammatory reaction involving both peripheral leukocytes and brain resident cells. Recent evidence indicates that their differentiation into a variety of functional phenotypes contributes to both tissue injury and repair. However, the temporal dynamics and diversity of post-stroke immune cell subsets remain poorly understood. To address these limitations, we performed a longitudinal single-cell transcriptomic study of both brain and mouse blood to obtain a composite picture of brain-infiltrating leukocytes, circulating leukocytes, microglia and endothelium diversity over the ischemic/reperfusion time. Brain cells and blood leukocytes isolated from mice 2 or 14 days after transient middle cerebral artery occlusion or sham surgery were purified by FACS sorting and processed for droplet-based single-cell transcriptomics. The analysis revealed a strong divergence of post-ischemic microglia, macrophages, and neutrophils over time, while such diversity was less evident in dendritic cells, B, T and NK cells. Conversely, brain endothelial cells and brain associated-macrophages showed altered transcriptomic signatures at 2 days post-stroke, but low divergence from sham at day 14. Pseudotime trajectory inference predicted the in-situ longitudinal progression of monocyte-derived macrophages from their blood precursors into day 2 and day 14 phenotypes, while microglia phenotypes at these two time points were not connected. In contrast to monocyte-derived macrophages, neutrophils were predicted to be continuously de-novo recruited from the blood. Brain single-cell transcriptomics from both female and male aged mice did not show major changes in respect to young mice, but aged and young brains differed in their immune cell composition. Furthermore, blood leukocyte analysis also revealed altered transcriptomes after stroke. However, brain-infiltrating leukocytes displayed higher transcriptomic divergence than their circulating counterparts, indicating that phenotypic diversification into cellular subsets occurs within the brain in the early and the recovery phase of ischemic stroke. In addition, this resource report contains a searchable database https://anratherlab.shinyapps.io/strokevis/ to allow user-friendly access to our data. The StrokeVis tool constitutes a comprehensive gene expression atlas that can be interrogated at the gene and cell type level to explore the transcriptional changes of endothelial and immune cell subsets from mouse brain and blood after stroke.
Collapse
Affiliation(s)
- Lidia Garcia-Bonilla
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Ziasmin Shahanoor
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Rose Sciortino
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Omina Nazarzoda
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Gianfranco Racchumi
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Costantino Iadecola
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Josef Anrather
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| |
Collapse
|
87
|
Jufar AH, Evans RG, May CN, Hood SG, Betrie AH, Trask‐Marino A, Bellomo R, Lankadeva YR. The effects of recruitment of renal functional reserve on renal cortical and medullary oxygenation in non-anesthetized sheep. Acta Physiol (Oxf) 2023; 237:e13919. [PMID: 36598336 PMCID: PMC10909474 DOI: 10.1111/apha.13919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/18/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
AIM Recruitment of renal functional reserve (RFR) with amino acid loading increases renal blood flow and glomerular filtration rate. However, its effects on renal cortical and medullary oxygenation have not been determined. Accordingly, we tested the effects of recruitment of RFR on renal cortical and medullary oxygenation in non-anesthetized sheep. METHODS Under general anesthesia, we instrumented 10 sheep to enable subsequent continuous measurements of systemic and renal hemodynamics, renal oxygen delivery and consumption, and cortical and medullary tissue oxygen tension (PO2 ). We then measured the effects of recruitment of RFR with an intravenous infusion of 500 ml of a clinically used amino acid solution (10% Synthamin® 17) in the non-anesthetized state. RESULTS Compared with baseline, Synthamin® 17 infusion significantly increased renal oxygen delivery mean ± SD maximum increase: (from 0.79 ± 0.17 to 1.06 ± 0.16 ml/kg/min, p < 0.001), renal oxygen consumption (from 0.08 ± 0.01 to 0.15 ± 0.02 ml/kg/min, p < 0.001), and glomerular filtration rate (+45.2 ± 2.7%, p < 0.001). Renal cortical tissue PO2 increased by a maximum of 26.4 ± 1.1% (p = 0.001) and medullary tissue PO2 increased by a maximum of 23.9 ± 2.8% (p = 0. 001). CONCLUSIONS In non-anesthetized healthy sheep, recruitment of RFR improved renal cortical and medullary oxygenation. These observations might have implications for the use of recruitment of RFR for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Alemayehu H. Jufar
- Pre‐Clinical Critical Care UnitFlorey Institute of Neuroscience and Mental Health, University of MelbourneMelbourneVictoriaAustralia
- Cardiovascular Disease Program, Department of PhysiologyBiomedicine Discovery Institute, Monash UniversityMelbourneVictoriaAustralia
| | - Roger G. Evans
- Pre‐Clinical Critical Care UnitFlorey Institute of Neuroscience and Mental Health, University of MelbourneMelbourneVictoriaAustralia
- Cardiovascular Disease Program, Department of PhysiologyBiomedicine Discovery Institute, Monash UniversityMelbourneVictoriaAustralia
| | - Clive N. May
- Pre‐Clinical Critical Care UnitFlorey Institute of Neuroscience and Mental Health, University of MelbourneMelbourneVictoriaAustralia
- Department of Critical CareMelbourne Medical School, University of MelbourneMelbourneVictoriaAustralia
| | - Sally G. Hood
- Pre‐Clinical Critical Care UnitFlorey Institute of Neuroscience and Mental Health, University of MelbourneMelbourneVictoriaAustralia
| | - Ashenafi H. Betrie
- Pre‐Clinical Critical Care UnitFlorey Institute of Neuroscience and Mental Health, University of MelbourneMelbourneVictoriaAustralia
- Melbourne Dementia Research CentreFlorey Institute of Neuroscience and Mental Health, The University of MelbourneMelbourneVictoriaAustralia
| | - Anton Trask‐Marino
- Pre‐Clinical Critical Care UnitFlorey Institute of Neuroscience and Mental Health, University of MelbourneMelbourneVictoriaAustralia
| | - Rinaldo Bellomo
- Pre‐Clinical Critical Care UnitFlorey Institute of Neuroscience and Mental Health, University of MelbourneMelbourneVictoriaAustralia
- Department of Critical CareMelbourne Medical School, University of MelbourneMelbourneVictoriaAustralia
| | - Yugeesh R. Lankadeva
- Pre‐Clinical Critical Care UnitFlorey Institute of Neuroscience and Mental Health, University of MelbourneMelbourneVictoriaAustralia
- Department of Critical CareMelbourne Medical School, University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
88
|
Chen X, Lu W, Lu C, Zhang L, Xu F, Dong H. The CaSR/TRPV4 coupling mediates pro-inflammatory macrophage function. Acta Physiol (Oxf) 2023; 237:e13926. [PMID: 36606511 DOI: 10.1111/apha.13926] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 12/29/2022] [Accepted: 01/02/2023] [Indexed: 01/07/2023]
Abstract
AIM Although calcium-sensing receptor (CaSR) and transient receptor potential vanilloid 4 (TRPV4) channels are functionally expressed on macrophages, it is unclear if they work coordinately to mediate macrophage function. The present study investigates whether CaSR couples to TRPV4 channels and mediates macrophage polarization via Ca2+ signaling. METHODS The role of CaSR/TRPV4/Ca2+ signaling was assessed in lipopolysaccharide (LPS)-treated peritoneal macrophages (PMs) from wild-type (WT) and TRPV4 knockout (TRPV4 KO) mice. The expression and function of CaSR and TRPV4 in PMs were analyzed by immunofluorescence and digital Ca2+ imaging. The correlation factors of M1 polarization, CCR7, IL-1β, and TNFα were detected using q-PCR, western blot, and ELISA. RESULTS We found that PMs expressed CaSR and TRPV4, and CaSR activation-induced marked Ca2+ signaling predominately through extracellular Ca2+ entry, which was inhibited by selective pharmacological blockers of CaSR and TRPV4 channels. The CaSR activation-induced Ca2+ signaling was significantly attenuated in PMs from TRPV4 KO mice compared to those from WT mice. Moreover, the CaSR activation-induced Ca2+ entry via TRPV4 channels was inhibited by blocking phospholipases A2 (PLA2)/cytochromeP450 (CYP450) and phospholipase C (PLC)/Protein kinase C (PKC) pathways. Finally, CaSR activation promoted the expression and release of M1-associated cytokines IL-1β and TNFɑ, which were attenuated in PMs from TRPV4 KO mice. CONCLUSION We reveal a novel coupling of the CaSR and TRPV4 channels via PLA2/CYP450 and PLC/PKC pathways, promoting a Ca2+ -dependent M1 macrophage polarization. Modulation of this coupling and downstream pathways may become a potential strategy for the prevention/treatment of immune-related disease.
Collapse
Affiliation(s)
- Xiongying Chen
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Lu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Cheng Lu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Luyun Zhang
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Feng Xu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Dong
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
89
|
Hu M, Zhang Y, Lu Y, Han J, Guo T, Cui P, Brännström M, Shao LR, Billig H. Regulatory mechanisms of HMGB1 and its receptors in polycystic ovary syndrome-driven gravid uterine inflammation. FEBS J 2023; 290:1874-1906. [PMID: 36380688 PMCID: PMC10952262 DOI: 10.1111/febs.16678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/23/2022] [Accepted: 11/15/2022] [Indexed: 11/17/2022]
Abstract
High-mobility group box 1 (HMGB1) is critical for inflammatory homeostasis and successful pregnancy, and there is a strong association among elevated levels of HMGB1, polycystic ovary syndrome (PCOS), chronic inflammation and pregnancy loss. However, the mechanisms responsible for PCOS-driven regulation of uterine HMGB1 and its candidate receptors [toll-like receptor (TLR) 2 and 4] and inflammatory responses during pregnancy remain unclear. In this study, we found a gestational stage-dependent decrease in uterine HMGB1 and TLR4 protein abundance in rats during normal pregnancy. We demonstrated that increased expression of HMGB1, TLR2 and TLR4 proteins was associated with activation of inflammation-related signalling pathways in the gravid uterus exposed to 5α-dihydrotestosterone and insulin, mimicking the clinical features (hyperandrogenism and insulin resistance) of PCOS and this elevation was completely inhibited by treatment with the androgen receptor (AR) antagonist flutamide. Interestingly, acute exposure to lipopolysaccharide suppressed HMGB1, TLR4 and inflammation-related protein abundance but did not affect androgen levels or AR expression in the gravid uterus with viable fetuses. Furthermore, immunohistochemical analysis revealed that, in addition to being localized predominately in the nuclear compartment, HMGB1 immunoreactivity was also detected in the cytoplasm in the PCOS-like rat uterus, PCOS endometrium and pregnant rat uterus with haemorrhagic and resorbed fetuses, possibly via activation of nuclear factor κB signalling. These results suggest that both AR-dependent and AR-independent mechanisms contribute to the modulation of HMGB1/TLR2/TLR4-mediated uterine inflammation. We propose that the elevation of HMGB1 and its receptors and disruption of the pro-/anti-inflammatory balance in the gravid uterus may participate in the pathophysiology of PCOS-associated pregnancy loss.
Collapse
Affiliation(s)
- Min Hu
- Department of Traditional Chinese MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityChina
- Institute of Integrated Traditional Chinese Medicine and Western MedicineGuangzhou Medical UniversityChina
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgSweden
| | - Yuehui Zhang
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgSweden
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated HospitalHeilongjiang University of Chinese MedicineHarbinChina
| | - Yaxing Lu
- Department of Traditional Chinese MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityChina
- Institute of Integrated Traditional Chinese Medicine and Western MedicineGuangzhou Medical UniversityChina
| | - Jing Han
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated HospitalHeilongjiang University of Chinese MedicineHarbinChina
| | - Tingting Guo
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated HospitalHeilongjiang University of Chinese MedicineHarbinChina
| | - Peng Cui
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgSweden
- Department of Obstetrics and GynecologyShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineChina
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Sahlgrenska AcademyUniversity of GothenburgSweden
| | - Linus R. Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgSweden
| | - Håkan Billig
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgSweden
| |
Collapse
|
90
|
Diong J, Bye E, Djajadikarta Z, Butler AA, Gandevia SC, Héroux ME. Encouraging responsible reporting practices in the Instructions to Authors of neuroscience and physiology journals: There is room to improve. PLoS One 2023; 18:e0283753. [PMID: 36996120 PMCID: PMC10062619 DOI: 10.1371/journal.pone.0283753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
Journals can substantially influence the quality of research reports by including responsible reporting practices in their Instructions to Authors. We assessed the extent to which 100 journals in neuroscience and physiology required authors to report methods and results in a rigorous and transparent way. For each journal, Instructions to Authors and any referenced reporting guideline or checklist were downloaded from journal websites. Twenty-two questions were developed to assess how journal Instructions to Authors address fundamental aspects of rigor and transparency in five key reporting areas. Journal Instructions to Authors and all referenced external guidelines and checklists were audited against these 22 questions. Of the full sample of 100 Instructions to Authors, 34 did not reference any external reporting guideline or checklist. Reporting whether clinical trial protocols were pre-registered was required by 49 journals and encouraged by 7 others. Making data publicly available was encouraged by 64 journals; making (processing or statistical) code publicly available was encouraged by ∼30 of the journals. Other responsible reporting practices were mentioned by less than 20 of the journals. Journals can improve the quality of research reports by mandating, or at least encouraging, the responsible reporting practices highlighted here.
Collapse
Affiliation(s)
- Joanna Diong
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia
| | - Elizabeth Bye
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | | | - Annie A. Butler
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Simon C. Gandevia
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia
- Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Martin E. Héroux
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
91
|
Interannual variability in early life phenology is driven by climate and oceanic processes in two NE Atlantic flatfishes. Sci Rep 2023; 13:4057. [PMID: 36906628 PMCID: PMC10008569 DOI: 10.1038/s41598-023-30384-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 02/21/2023] [Indexed: 03/13/2023] Open
Abstract
Early life phenology is a crucial factor for population dynamics in a climate change scenario. As such, understanding how the early life cycle of marine fishes is influenced by key oceanic and climate drivers is of chief importance for sustainable fisheries. This study documents interannual changes in early life phenology of two commercial flatfishes: European flounder (Platichthys flesus) and common sole (Solea solea) from 2010 to 2015 based on otolith microstructure. Using GAMs, we looked for correlations of the North Atlantic Oscillation (NAO), Eastern Atlantic pattern (EA), sea surface temperature (SST), chlorophyl a concentration (Chla) and upwelling (Ui) variation with the onset of hatch, metamorphosis, and benthic settlement day. We concluded that higher SST, more intensive upwelling, and EA were coincident with a later the onset of each stage, while increasing NAO induces an earlier onset of each stage. Although similar to S. solea, P. flesus showed a more complex interaction with the environmental drivers, most possibly because it is at its southern limit of its distribution. Our results highlight the complexity of the relationship between climate conditions and fish early life history, particularly those with complex life cycles that include migrations between coastal areas and estuaries.
Collapse
|
92
|
Guo Y, Zhu H, Wang Y, Sun T, Xu J, Wang T, Guan W, Wang C, Liu C, Ma C. Miniature-swine iPSC-derived GABA progenitor cells function in a rat Parkinson's disease model. Cell Tissue Res 2023; 391:425-440. [PMID: 36645476 DOI: 10.1007/s00441-022-03736-4] [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: 03/01/2022] [Accepted: 12/20/2022] [Indexed: 01/17/2023]
Abstract
Induced pluripotent stem cells (iPS cells) are considered a promising source of cell-based therapy for the treatment of Parkinson's disease (PD). Recent studies have shown forebrain GABA interneurons have crucial roles in many psychiatric disorders, and secondary changes in the GABA system play a directly effect on the pathogenesis of PD. Here, we first describe an efficient differentiation procedure of GABA progenitors (MiPSC-iGABAPs) from miniature-swine iPSCs through two major developmental stages. Then, the MiPSC-iGABAPs were stereotactically transplanted into the right medial forebrain bundle (MFB) of 6-hydroxydopamine (OHDA)-lesioned PD model rats to confirm their feasibility for the neural transplantation as a donor material. Furthermore, the grafted MiPSC-iGABAPs could survive and migrate from the graft site into the surrounding brain tissue including striatum (ST) and substantia nigra (SN) for at least 32 weeks, and significantly improved functional recovery of PD rats from their parkinsonian behavioral defects. Histological studies showed that the grafted cells could migrate and differentiate into various neurocytes, including GABAergic, dopaminergic neurons, and glial cells in vivo, and many induced dopaminergic neurons extended dense neurites into the host striatum. Moreover, over 50% of the grafted MiPSC-iGABAPs could express GABA, and these GABAergic neurons might be responsible for modifying the balance of excitatory and inhibitory signals in the striatum to promote behavioral recovery. Thus, the present study confirmed that the MiPSC-iGABAPs can be used as an attractive donor material for the neural grafting to remodel basal ganglia circuitry in neurodegenerative diseases, avoiding tumorigenicity of iPSCs and the nonproliferative and nondifferentiated potential of mature neurons.
Collapse
Affiliation(s)
- Yu Guo
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Huan Zhu
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Yuanyuan Wang
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Tingting Sun
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Jiajia Xu
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Tie Wang
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Weijun Guan
- Institute of Beijing Animal Science and Veterinary, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Chunjing Wang
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Changqing Liu
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China. .,Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| | - Caiyun Ma
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
93
|
Wang XY, Chen XQ, Wang GQ, Cai RL, Wang H, Wang HT, Peng XQ, Zhang MT, Huang S, Shen GM. A neural circuit for gastric motility disorders driven by gastric dilation in mice. Front Neurosci 2023; 17:1069198. [PMID: 36908796 PMCID: PMC9992744 DOI: 10.3389/fnins.2023.1069198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
INTRODUCTION Symptoms of gastric motility disorders are common clinical manifestations of functional gastrointestinal disorders (FGIDs), and are triggered and exacerbated by stress, but the neural pathways underpinning them remain unclear. METHODS We set-up a mouse model by gastric dilation (GD) in which the gastric dynamics were assessed by installing strain gauges on the surface of the stomach. The neural pathway associated with gastric motility disorders was investigated by behavioral tests, electrophysiology, neural circuit tracing, and optogenetics and chemogenetics involving projections of the corticotropin-releasing hormone (CRH) from the paraventricular nucleus of the hypothalamus (PVN) to acetylcholine (ChAT) neurons in the dorsal motor nucleus of the vagus (DMV). RESULTS We found that GD induced gastric motility disorders were accompanied by activation of PVN CRH neurons, which could be alleviated by strategies that inhibits the activity of PVN CRH neurons. In addition, we identified a neural pathway in which PVN CRH neurons project into DMV ChAT neurons, modulated activity of the PVN CRH →DMV ChAT pathway to alleviate gastric motility disorders induced by GD. DISCUSSION These findings indicate that the PVN CRH →DMV ChAT pathway may mediate at least some aspects of GD related gastric motility, and provide new insights into the mechanisms by which somatic stimulation modulates the physiological functions of internal organs and systems.
Collapse
Affiliation(s)
- Xi-yang Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiao-qi Chen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Guo-quan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Rong-lin Cai
- Research Institute of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hao Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hai-tao Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiao-qi Peng
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Meng-ting Zhang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Shun Huang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Guo-ming Shen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Institute of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
94
|
Lear CA, Georgieva A, Beacom MJ, Wassink G, Dhillon SK, Lear BA, Mills OJ, Westgate JA, Bennet L, Gunn AJ. Fetal heart rate responses in chronic hypoxaemia with superimposed repeated hypoxaemia consistent with early labour: a controlled study in fetal sheep. BJOG 2023. [PMID: 36808862 DOI: 10.1111/1471-0528.17425] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/20/2023]
Abstract
OBJECTIVE Deceleration area (DA) and capacity (DC) of the fetal heart rate can help predict risk of intrapartum fetal compromise. However, their predictive value in higher risk pregnancies is unclear. We investigated whether they can predict the onset of hypotension during brief hypoxaemia repeated at a rate consistent with early labour in fetal sheep with pre-existing hypoxaemia. DESIGN Prospective, controlled study. SETTING Laboratory. SAMPLE Chronically instrumented, unanaesthetised near-term fetal sheep. METHODS One-minute complete umbilical cord occlusions (UCOs) were performed every 5 minutes in fetal sheep with baseline pa O2 <17 mmHg (hypoxaemic, n = 8) and >17 mmHg (normoxic, n = 11) for 4 hours or until arterial pressure fell <20 mmHg. MAIN OUTCOME MEASURES DA, DC and arterial pressure. RESULTS Normoxic fetuses showed effective cardiovascular adaptation without hypotension and mild acidaemia (lowest arterial pressure 40.7 ± 2.8 mmHg, pH 7.35 ± 0.03). Hypoxaemic fetuses developed hypotension (lowest arterial pressure 20.8 ± 1.9 mmHg, P < 0.001) and acidaemia (final pH 7.07 ± 0.05). In hypoxaemic fetuses, decelerations showed faster falls in FHR over the first 40 seconds of UCOs but the final deceleration depth was not different to normoxic fetuses. DC was modestly higher in hypoxaemic fetuses during the penultimate (P = 0.04) and final (P = 0.012) 20 minutes of UCOs. DA was not different between groups. CONCLUSION Chronically hypoxaemic fetuses had early onset of cardiovascular compromise during labour-like brief repeated UCOs. DA was unable to identify developing hypotension in this setting, while DC only showed modest differences between groups. These findings highlight that DA and DC thresholds need to be adjusted for antenatal risk factors, potentially limiting their clinical utility.
Collapse
Affiliation(s)
- C A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - A Georgieva
- Nuffield Department of Women's and Reproductive Health, The John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - M J Beacom
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - G Wassink
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - S K Dhillon
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - B A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - O J Mills
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - J A Westgate
- Department of Obstetrics and Gynaecology, The University of Auckland, Auckland, New Zealand
| | - L Bennet
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - A J Gunn
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand.,Starship Children's Hospital, Auckland, New Zealand
| |
Collapse
|
95
|
Vercalsteren E, Karampatsi D, Dekens D, Letsiou A, Zabala A, Romanitan M, Klein T, Nyström T, Darsalia V, Patrone C. The Pre-Stroke Induction and Normalization of Insulin Resistance Respectively Worsens and Improves Functional Recovery. Int J Mol Sci 2023; 24:ijms24043989. [PMID: 36835405 PMCID: PMC9964646 DOI: 10.3390/ijms24043989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
Type 2 diabetes (T2D) impairs post-stroke recovery, and the underlying mechanisms are unknown. Insulin resistance (IR), a T2D hallmark that is also closely linked to aging, has been associated with impaired post-stroke recovery. However, whether IR worsens stroke recovery is unknown. We addressed this question in mouse models where early IR, with or without hyperglycemia, was induced by chronic high-fat diet feeding or sucrose supplementation in the drinking water, respectively. Furthermore, we used 10-month-old mice, spontaneously developing IR but not hyperglycemia, where IR was normalized pharmacologically pre-stroke with Rosiglitazone. Stroke was induced by transient middle cerebral artery occlusion and recovery was assessed by sensorimotor tests. Neuronal survival, neuroinflammation and the density of striatal cholinergic interneurons were also assessed by immunohistochemistry/quantitative microscopy. Pre-stroke induction and normalization of IR, respectively, worsened and improved post-stroke neurological recovery. Moreover, our data indicate a potential association of this impaired recovery with exacerbated neuroinflammation and a decreased density of striatal cholinergic interneurons. The global diabetes epidemic and population aging are dramatically increasing the percentage of people in need of post-stroke treatment/care. Our results suggest that future clinical studies should target pre-stroke IR to reduce stroke sequelae in both diabetics and elderly people with prediabetes.
Collapse
Affiliation(s)
- Ellen Vercalsteren
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83 Stockholm, Sweden
| | - Dimitra Karampatsi
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83 Stockholm, Sweden
| | - Doortje Dekens
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83 Stockholm, Sweden
| | - Aikaterini Letsiou
- Neurology Department, Internal Medicine, Södersjukhuset, 118 83 Stockholm, Sweden
| | - Alexander Zabala
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83 Stockholm, Sweden
| | - Mihaela Romanitan
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83 Stockholm, Sweden
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co., KG, 88400 Biberach, Germany
| | - Thomas Nyström
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83 Stockholm, Sweden
| | - Vladimer Darsalia
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83 Stockholm, Sweden
- Correspondence: (V.D.); (C.P.); Tel.: +46-(8)-6165084 (V.D. & C.P.); Fax: +46-(8)-6162933 (V.D.); +46-(8)-6162933 (C.P.)
| | - Cesare Patrone
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83 Stockholm, Sweden
- Correspondence: (V.D.); (C.P.); Tel.: +46-(8)-6165084 (V.D. & C.P.); Fax: +46-(8)-6162933 (V.D.); +46-(8)-6162933 (C.P.)
| |
Collapse
|
96
|
Sahu SK, Ozantürk AN, Kulkarni DH, Ma L, Barve RA, Dannull L, Lu A, Starick M, McPhatter J, Garnica L, Sanfillipo-Burchman M, Kunen J, Wu X, Gelman AE, Brody SL, Atkinson JP, Kulkarni HS. Lung epithelial cell-derived C3 protects against pneumonia-induced lung injury. Sci Immunol 2023; 8:eabp9547. [PMID: 36735773 PMCID: PMC10023170 DOI: 10.1126/sciimmunol.abp9547] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023]
Abstract
The complement component C3 is a fundamental plasma protein for host defense, produced largely by the liver. However, recent work has demonstrated the critical importance of tissue-specific C3 expression in cell survival. Here, we analyzed the effects of local versus peripheral sources of C3 expression in a model of acute bacterial pneumonia induced by Pseudomonas aeruginosa. Whereas mice with global C3 deficiency had severe pneumonia-induced lung injury, those deficient only in liver-derived C3 remained protected, comparable to wild-type mice. Human lung transcriptome analysis showed that secretory epithelial cells, such as club cells, express high levels of C3 mRNA. Mice with tamoxifen-induced C3 gene ablation from club cells in the lung had worse pulmonary injury compared with similarly treated controls, despite maintaining normal circulating C3 levels. Last, in both the mouse pneumonia model and cultured primary human airway epithelial cells, we showed that stress-induced death associated with C3 deficiency parallels that seen in Factor B deficiency rather than C3a receptor deficiency. Moreover, C3-mediated reduction in epithelial cell death requires alternative pathway component Factor B. Thus, our findings suggest that a pathway reliant on locally derived C3 and Factor B protects the lung mucosal barrier.
Collapse
Affiliation(s)
- Sanjaya K. Sahu
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Ayşe N. Ozantürk
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Devesha H. Kulkarni
- Division of Gastroenterology, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Lina Ma
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Ruteja A Barve
- Department of Genetics, Washington University School of Medicine; St. Louis, USA
| | - Linus Dannull
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Angel Lu
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Marick Starick
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Ja’Nia McPhatter
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Lorena Garnica
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Maxwell Sanfillipo-Burchman
- Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine; St. Louis, USA
| | - Jeremy Kunen
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Xiaobo Wu
- Division of Rheumatology, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Andrew E. Gelman
- Department of Surgery, Washington University School of Medicine; St. Louis, USA
| | - Steven L. Brody
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - John P. Atkinson
- Division of Rheumatology, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| | - Hrishikesh S. Kulkarni
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine; St. Louis, USA
| |
Collapse
|
97
|
Walker AE, Cole JA, Krishna Kumaran S, Kato JI, Zhuang X, Wolf JR, Henson GD, McCully BH. INFLUENCE OF OBESITY ON VASCULAR DYSFUNCTION AFTER TRAUMATIC HEMORRHAGE. Shock 2023; 59:318-325. [PMID: 36731028 DOI: 10.1097/shk.0000000000001991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
ABSTRACT Background: Obesity increases the risk for morbidity and mortality after trauma. These complications are associated with profound vascular damage. Traumatic hemorrhage acutely attenuates vascular responsiveness, but the impact of obesity on this dysfunction is not known. The local inflammatory response in vascular cells is also unknown. We hypothesized that obesity potentiates trauma-induced vascular inflammation and dysfunction. Methods: Male Sprague-Dawley rats (~250 g) were fed normal chow (NC; 13.5% kcal fat, n = 20) or high-fat (HF; 60% kcal fat, n = 20) diets for 6 to 8 weeks. Under anesthesia, hemorrhage was induced by a mesenteric artery laceration, a Grade V splenic injury, and hypotension (MAP = 30-40 mm Hg) for 30 minutes. Vascular responsiveness was assessed ex vivo in isolated mesenteric arteries prehemorrhage and posthemorrhage. Gene expression for IL-1β, and IL-6, prooxidant nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2), and α-adrenergic receptor were assessed in carotid artery endothelial cells (ECs) and non-ECs (media + adventitia). Results: In NC rats, hemorrhage attenuated norepinephrine-induced vasoconstriction and endothelium-dependent vasodilation to acetylcholine. In HF rats, baseline norepinephrine-induced vasoconstriction was attenuated compared with NC, but vasoconstriction and endothelium-dependent vasodilation did not change prehemorrhage to posthemorrhage. Hemorrhage led to elevated IL-1β gene expression in ECs and elevated IL1β, IL-6, NOX2, and α-adrenergic receptor gene expression in the media + adventitia compared with sham. HF rats had greater EC IL-1 β and NOX2 gene expression compared with NC rats. The hemorrhage-induced elevation of IL-1β in the media + adventitia was greatest in HF rats. Conclusion: Traumatic hemorrhage attenuates vascular responsiveness and induces vascular inflammation. The attenuated vascular responsiveness after hemorrhage is absent in obese rats, while the elevated vascular inflammation persists. A HF diet amplifies the arterial inflammation after hemorrhage. Altered vascular responsiveness and vascular inflammation may contribute to worse outcomes in obese trauma patients.
Collapse
Affiliation(s)
- Ashley E Walker
- Department of Human Physiology, University of Oregon, Eugene, OR
| | - Jazmin A Cole
- Department of Human Physiology, University of Oregon, Eugene, OR
| | | | - Jonathan I Kato
- Department of Human Physiology, University of Oregon, Eugene, OR
| | - Xinhao Zhuang
- Department of Human Physiology, University of Oregon, Eugene, OR
| | - Julia R Wolf
- Department of Human Physiology, University of Oregon, Eugene, OR
| | - Grant D Henson
- Department of Human Physiology, University of Oregon, Eugene, OR
| | | |
Collapse
|
98
|
Monge García MI, Jian Z, Hatib F, Settles JJ, Cecconi M, Pinsky MR. Relationship between intraventricular mechanical dyssynchrony and left ventricular systolic and diastolic performance: An in vivo experimental study. Physiol Rep 2023; 11:e15607. [PMID: 36808901 PMCID: PMC9937795 DOI: 10.14814/phy2.15607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/13/2023] [Accepted: 01/23/2023] [Indexed: 02/19/2023] Open
Abstract
Left ventricular mechanical dyssynchrony (LVMD) refers to the nonuniformity in mechanical contraction and relaxation timing in different ventricular segments. We aimed to determine the relationship between LVMD and LV performance, as assessed by ventriculo-arterial coupling (VAC), LV mechanical efficiency (LVeff ), left ventricular ejection fraction (LVEF), and diastolic function during sequential experimental changes in loading and contractile conditions. Thirteen Yorkshire pigs submitted to three consecutive stages with two opposite interventions each: changes in afterload (phenylephrine/nitroprusside), preload (bleeding/reinfusion and fluid bolus), and contractility (esmolol/dobutamine). LV pressure-volume data were obtained with a conductance catheter. Segmental mechanical dyssynchrony was assessed by global, systolic, and diastolic dyssynchrony (DYS) and internal flow fraction (IFF). Late systolic LVMD was related to an impaired VAC, LVeff , and LVEF, whereas diastolic LVMD was associated with delayed LV relaxation (logistic tau), decreased LV peak filling rate, and increased atrial contribution to LV filling. The hemodynamic factors related to LVMD were contractility, afterload, and heart rate. However, the relationship between these factors differed throughout the cardiac cycle. LVMD plays a significant role in LV systolic and diastolic performance and is associated with hemodynamic factors and intraventricular conduction.
Collapse
Affiliation(s)
| | | | | | | | - Maurizio Cecconi
- Department Anaesthesia and Intensive Care Units, Humanitas Research HospitalHumanitas UniversityMilanItaly
| | - Michael R. Pinsky
- Department of Critical Care MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
99
|
Dhillon SK, Gunn ER, Pedersen MV, Lear CA, Wassink G, Davidson JO, Gunn AJ, Bennet L. Alpha-adrenergic receptor activation after fetal hypoxia-ischaemia suppresses transient epileptiform activity and limits loss of oligodendrocytes and hippocampal neurons. J Cereb Blood Flow Metab 2023; 43:947-961. [PMID: 36703575 DOI: 10.1177/0271678x231153723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Exposure to hypoxic-ischaemia (HI) is consistently followed by a delayed fall in cerebral perfusion. In preterm fetal sheep this is associated with impaired cerebral oxygenation, consistent with mismatch between perfusion and metabolism. In the present study we tested the hypothesis that alpha-adrenergic inhibition after HI would improve cerebral perfusion, and so attenuate mismatch and reduce neural injury. Chronically instrumented preterm (0.7 gestation) fetal sheep received sham-HI (n = 10) or HI induced by complete umbilical cord occlusion for 25 minutes. From 15 minutes to 8 hours after HI, fetuses received either an intravenous infusion of a non-selective alpha-adrenergic antagonist, phentolamine (10 mg bolus, 10 mg/h infusion, n = 10), or saline (n = 10). Fetal brains were processed for histology 72 hours post-HI. Phentolamine infusion was associated with increased epileptiform transient activity and a greater fall in cerebral oxygenation in the early post-HI recovery phase. Histologically, phentolamine was associated with greater loss of oligodendrocytes and hippocampal neurons. In summary, contrary to our hypothesis, alpha-adrenergic inhibition increased epileptiform transient activity with an exaggerated fall in cerebral oxygenation, and increased neural injury, suggesting that alpha-adrenergic receptor activation after HI is an important endogenous neuroprotective mechanism.
Collapse
Affiliation(s)
| | - Eleanor R Gunn
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Mette V Pedersen
- Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark
| | - Christopher A Lear
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Guido Wassink
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Joanne O Davidson
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
100
|
Li L, Li P, Guo J, Wu Y, Zeng Q, Li N, Huang X, He Y, Ai W, Sun W, Liu T, Xiong D, Xiao L, Sun Y, Zhou Q, Kuang H, Wang Z, Jiang C. Up-regulation of oxytocin receptors on peripheral sensory neurons mediates analgesia in chemotherapy-induced neuropathic pain. Br J Pharmacol 2023. [PMID: 36702458 DOI: 10.1111/bph.16042] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 01/19/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND PURPOSE Chemotherapy-induced neuropathic pain (CINP) currently has limited effective treatment. Although the roles of oxytocin (OXT) and the oxytocin receptor (OXTR) in central analgesia have been well documented, the expression and function of OXTR in the peripheral nervous system remain unclear. Here, we evaluated the peripheral antinociceptive profiles of OXTR in CINP. EXPERIMENTAL APPROACH Paclitaxel (PTX) was used to establish CINP. Quantitative real-time polymerase chain reaction (qRT-PCR), in situ hybridization, and immunohistochemistry were used to observe OXTR expression in dorsal root ganglia (DRG). The antinociceptive effects of OXT were assessed by hot-plate and von Frey tests. Whole-cell patch clamp was performed to record sodium currents, excitability of DRG neurons, and excitatory synapse transmission. KEY RESULTS Expression of OXTR in DRG neurons was enhanced significantly after PTX treatment. Activation of OXTR exhibited antinociceptive effects, by decreasing the hyperexcitability of DRG neurons in PTX-treated mice. Additionally, OXTR activation up-regulated the phosphorylation of protein kinase C (pPKC) and, in turn, impaired voltage-gated sodium currents, particularly the voltage-gated sodium channel 1.7 (NaV 1.7) current, that plays an indispensable role in PTX-induced neuropathic pain. OXT suppressed excitatory transmission in the spinal dorsal horn as well as excitatory inputs from primary afferents in PTX-treated mice. CONCLUSION AND IMPLICATIONS The OXTR in small-sized DRG neurons is up-regulated in CINP and its activation relieved CINP by inhibiting the neural excitability by impairment of NaV 1.7 currents via pPKC. Our results suggest that OXTR on peripheral sensory neurons is a potential therapeutic target to relieve CINP.
Collapse
Affiliation(s)
- Lixuan Li
- Guangdong Medical University, Zhanjiang, Guangdong, China.,Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Pupu Li
- Department of Medical Oncology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Jing Guo
- Department of Endocrinology and Metabolism, Shenzhen University General Hospital and Shenzhen University Academy of Clinical Medical Sciences, Shenzhen University, Shenzhen, Guangdong, China
| | - Yifei Wu
- Department of Medical Neuroscience, Key University Laboratory of Metabolism and Health of Guangdong, SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Qian Zeng
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Nan Li
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Xiaoting Huang
- Medical Research Center, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Yongshen He
- Medical Research Center, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Wen Ai
- Medical Research Center, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Wuping Sun
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Tao Liu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Donglin Xiong
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Lizu Xiao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Yanyan Sun
- Department of Anesthesiology, Shenzhen University General Hospital and Shenzhen University Academy of Clinical Medical Sciences, Shenzhen University, Shenzhen, Guangdong, China
| | - Qiming Zhou
- Department of Medical Oncology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Haixia Kuang
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zilong Wang
- Department of Medical Neuroscience, Key University Laboratory of Metabolism and Health of Guangdong, SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Changyu Jiang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China.,Medical Research Center, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| |
Collapse
|