You are on page 1of 67

Oxford textbook of neuroscience and

anaesthesiology First Edition. Edition


Engelhard
Visit to download the full and correct content document:
https://ebookmass.com/product/oxford-textbook-of-neuroscience-and-anaesthesiolog
y-first-edition-edition-engelhard/
i

Oxford Textbook of

Neuroscience and
Anaesthesiology
ii

Oxford Textbooks In Anaesthesia

Oxford Textbook of Anaesthesia for the Elderly Patient


Edited by Chris Dodds, Chandra M. Kumar, and Bernadette Th.Veering
Oxford Textbook of Anaesthesia for Oral and Maxillofacial Surgery
Edited by Ian Shaw, Chandra M. Kumar, and Chris Dodds
Principles and Practice of Regional Anaesthesia, Fourth Edition
Edited by Graeme McLeod, Colin McCartney, and Tony Wildsmith
Oxford Textbook of Cardiothoracic Anaesthesia
Edited by R. Peter Alston, Paul S. Myles, and Marco Ranucci
Oxford Textbook of Transplant Anaesthesia and Critical Care
Edited by Ernesto A. Pretto, Jr., Gianni Biancofiore, Andre DeWolf, John R. Klinck, Claus Niemann,
Andrew Watts, and Peter D. Slinger
Oxford Textbook of Obstetric Anaesthesia
Edited by Vicki Clark, Marc Van de Velde, Roshan Fernando
Oxford Textbook of Neuroscience and Anaesthesiology
Edited by George A. Mashour and Kristin Engelhard
iii

Oxford Textbook of

Neuroscience
and
Anaesthesiology
Edited by
George A. Mashour
Bert N. La Du Professor of Anesthesiology Research
Professor of Anesthesiology and Neurosurgery
Faculty, Neuroscience Graduate Program
Director, Center for Consciousness Science
Director, Michigan Institute for Clinical & Health Research
Associate Dean for Clinical and Translational Research
University of Michigan Medical School
Ann Arbor, Michigan, USA

Kristin Engelhard
Professor of Anesthesiology
Vice-​Chair of the Department of Anesthesiology
University Medical Center of the Johannes Gutenberg-​University
Mainz, Germany

1
iv

1
Great Clarendon Street, Oxford, OX2 6DP,
United Kingdom
Oxford University Press is a department of the University of Oxford.
It furthers the University’s objective of excellence in research, scholarship,
and education by publishing worldwide. Oxford is a registered trade mark of
Oxford University Press in the UK and in certain other countries
© Oxford University Press 2019
The moral rights of the authors have been asserted
First Edition published in 2019
Impression: 1
All rights reserved. No part of this publication may be reproduced, stored in
a retrieval system, or transmitted, in any form or by any means, without the
prior permission in writing of Oxford University Press, or as expressly permitted
by law, by licence or under terms agreed with the appropriate reprographics
rights organization. Enquiries concerning reproduction outside the scope of the
above should be sent to the Rights Department, Oxford University Press, at the
address above
You must not circulate this work in any other form
and you must impose this same condition on any acquirer
Published in the United States of America by Oxford University Press
198 Madison Avenue, New York, NY 10016, United States of America
British Library Cataloguing in Publication Data
Data available
Library of Congress Control Number: 2018947871
ISBN 978–​0–​19–​874664–​5
Printed in Great Britain by
Bell & Bain Ltd., Glasgow
Oxford University Press makes no representation, express or implied, that the
drug dosages in this book are correct. Readers must therefore always check
the product information and clinical procedures with the most up-​to-​date
published product information and data sheets provided by the manufacturers
and the most recent codes of conduct and safety regulations. The authors and
the publishers do not accept responsibility or legal liability for any errors in the
text or for the misuse or misapplication of material in this work. Except where
otherwise stated, drug dosages and recommendations are for the non-​pregnant
adult who is not breast-​feeding
Links to third party websites are provided by Oxford in good faith and
for information only. Oxford disclaims any responsibility for the materials
contained in any third party website referenced in this work.
v

Dedication

George A. Mashour Kristin Engelhard


Dedicated to my wonderful children, Alexander Fulgens Mashour Dedicated to my mentors and teachers Eberhard Kochs and Christian
and Anna Luise Mashour—​may they live long, healthy, and joyful Werner, who always encouraged and supported me throughout my
lives, and reach the fullest potential of their beautiful minds. academic career.
vi
vi

Preface-the three pillars


of Neuroanaesthesiology

While serving as the President of the Society for Neuroscience The Oxford Textbook of Neuroscience and Anaesthesiology is the
in Anesthesiology and Critical Care, I espoused a vision for first book of its kind to comprehensively address all three pillars
neuroanaesthesiology that was supported by three ‘pillars’. The related to neuroscience in anaesthesiology. The first section treats
traditional pillar of neuroanaesthesiology relates to the care of the neuroscientific foundations of anaesthesiology, including the
neurosurgical and neurological patients. The clinical care of indi- neural mechanisms of general anaesthetics, cerebral physiology,
viduals with neurologic compromise is incredibly rewarding and the neurobiology of pain, and more. The second section represents
represents a true opportunity to make a positive difference in the the traditional pillar related to the care of patients with neuro-
lives of others. However, the specialty of anaesthesiology is itself a logic disease in the operating room or intensive care unit, with a
form of clinical neuroscience. On a daily basis, even as anaesthe- focus on clinical neuroanaesthesia. These chapters systematically
tists for non-​neurosurgical cases, we modulate peripheral nerves, treat the peri-​operative considerations of both brain and spine
the spinal cord, subcortical arousal systems, thalamocortical and surgery, and provide introductions to neurocritical care and pedi-
corticocortical networks supporting consciousness, pain networks, atric neuroanaesthesia. Finally, the last section examines some
memory systems in the medial temporal lobe, the neuromuscular connections of neurology and anaesthesiology, examining how
junction, and the autonomic nervous system. From this perspective, conditions such as dementia, stroke, or epilepsy interface with the
‘neuroanaesthesiology’ is more a compression of ‘neuroscience in peri-​operative period.
anaesthesiology’ than ‘neurosurgical anaesthesiology’. The mech- This international textbook gathers the best available expertise of
anistic study of our therapeutic interventions, which represents authors and leaders in the field from Canada, Germany, Italy, New
another pillar, is exciting neuroscience in its own right, and has pro- Zealand, Spain, Switzerland, the UK, and the US. They have done an
found implications for nervous system function. Finally, the ques- outstanding job of crafting concise yet highly informative chapters
tion of how the peri-​operative period might negatively impact the describing the cutting edge of neuroscience and neuroanaesthesia.
brain is the new frontier of outcomes studies and has been a major It is my hope that this textbook is itself a ‘chapter’ in the evolution
priority for the field of anaesthesiology in the past decade. Questions of the field, creating a lasting foundation and appreciation for the
related to anaesthetic neurotoxicity, cognitive dysfunction, stroke, three pillars of neuroscience in anaesthesiology.
and other neurologic outcomes of non-​neurosurgical interventions
represent a critically important third pillar for the subspecialty. George A. Mashour, M.D., Ph.D.
vi
ix

Contents

Abbreviations xi 11 Neurophysiologic Monitoring


Contributors xv
for Neurosurgery 137
Antoun Koht, Laura B. Hemmer,
Digital media accompanying the book xvii J. Richard Toleikis, and Tod B. Sloan
12 Brain Trauma 149
SECTION 1​ Anne Sebastiani and Kristin Engelhard
Neuroscience in Anaesthetic Practice 13 Supratentorial Craniotomy for
1 Neural Mechanisms of Anaesthetics 3 Mass Lesion 161
Andrew McKinstry-​Wu and Max B. Kelz Shaun E. Gruenbaum and Federico Bilotta

2 Intracranial Pressure 17 14 The Posterior Fossa 173

Harald Stefanits, Andrea Reinprecht, Tasha L. Welch and Jeffrey J. Pasternak


and Klaus Ulrich Klein 15 Cerebrovascular Surgery 189
3 Cerebral Physiology 27 Deepak Sharma and David R. Wright
Stefan Bittner, Kerstin Göbel, and Sven G. Meuth 16 Interventional Neuroradiology 201
4 Introduction to Electroencephalography 35 Nathan Manning, Katherine M. Gelber, Michael
Michael Avidan and Jamie Sleigh Crimmins, Philip M. Meyers, and Eric J. Heyer

5 The Autonomic Nervous System 47 17 Pituitary and Neuroendocrine Surgery 213

David B. Glick, Gerald Glick†, and Erica J. Stein Douglas A. Colquhoun and Edward C. Nemergut

6 Neuromuscular Junction: Anatomy and 18 Hydrocephalus and Associated Surgery 225


Physiology, Paralytics, and Reversal Agents 61 Paola Hurtado and Neus Fàbregas
Christiane G. Stäuble, Heidrun Lewald, and Manfred Blobner 19 Awake Craniotomy for Tumour, Epilepsy,
7 Principles of Neuroprotection 77 and Functional Neurosurgery 235
Sophia C. Yi, Brian P. Lemkuil, and Piyush Patel Lashmi Venkatraghavan and Pirjo Manninen

8 Neurotoxicity of General Anaesthetics 93 20 Anaesthesia for Complex Spine Surgeries 245

Margaret K. Menzel Ellis and Ansgar Brambrink Ehab Farag and Zeyd Ebrahim

9 Neurobiology of Acute and Chronic Pain 111 21 Spine Trauma 255

Adrian Pichurko and Richard E. Harris Timur M. Urakov and Michael Y. Wang
22 Paediatric Neuroanaesthesia 263

SECTION 2​ Sulpicio G. Soriano and Craig D. McClain


Clinical Neuroanaesthesia 23 Basics of Neurocritical Care 273
Magnus Teig and Martin Smith
10 Neurologic Emergencies 125
Ross P. Martini and Ines P. Koerner
x

x  contents

SECTION 3​ 26 Epilepsy 303

Neurologic Patients Undergoing Adam D. Niesen, Adam K. Jacob, and Sandra L. Kopp
Non-​Neurologic Surgery 27 Parkinson’s Disease 309
M. Luke James and Ulrike Hoffmann
24 Cerebrovascular Disease 289
Corey Amlong and Robert D. Sanders 28 Treatment of Psychiatric Diseases
with General Anaesthetics 315
25 Peri-Operative Considerations of Dementia,
Laszlo Vutskits
Delirium, and Cognitive Decline 297
Phillip E. Vlisides and Zhongcong Xie
Index 323
xi

Abbreviations

133Xe Xenon BAER Brainstem auditory evoked response


3D Three-​dimensional BBB Blood-​brain barrier
AANS American Association of Neurological Surgeons BDNF Brain-​derived neurotrophic factor
ABC Airway, breathing, circulation BF Basal forebrain
ABCB-​1 ATP-​binding cassette subfamily B member 1 BIS Bispectral index
ABI Acute brain injury BIS Bispectral index
ABP Arterial blood pressure BK Bradykinin
ABR Auditory brain stem responses BP Blood pressure
ACA Anterior cerebral artery BTF Brain Trauma Foundation
ACC Anterior cingulate cortex Ca Aterial concentration
ACDF Anterior cervical discectomy with fusion cAMP Cyclic adenosine monophosphate
ACh Acetylcholine CAS Carotid artery stenosis
AChE acetylcholinesterase CAT-​1 Cationic amino-​acid transporter type 1
ACSNSQIP American College of Surgeons National Surgical CBF Cerebral blood flow
Quality Improvement Program CBV Cerebral blood volume
ACTH Adrenocorticotropic hormone CBVS Cerebrovascular surgery
ADH Antidiuretic hormone CCS Central cord syndrome
ADHD attention deficit hyperactivity disorder CCT Cranial computed tomography
AED Anti-​epileptic drug CEA Carotid endarterectomy
AION Anterior ischemic optic neuropathy CE-​MRC Contrast material-​enhanced MR cisternography
AIS Abbreviated Injury Scale CGRP Calcitonin gene-​related peptide
AIS Acute ischemic stroke CHD Congenital heart disease
AMPA α-​amino-​3-​hydroxy-​5-​methyl-​4-​ CHF Congestive heart failure
isoxazolepropionate CI Cardiac index
ANP Atrial natriuretic peptide CIC Intracerebral compliance
ANS Autonomic nervous system CM Cerebral microdialysis
AQP1 Aquaporin-​1 CMAP Compound muscle action potential
AQP4 Aquaporin-​4 CMR Cerebral metabolic rate
AQPs Aquaporins CMRO2 Cerebral metabolic oxygen consumption
ARAS Ascending reticular activating system CMT Central medial thalamus
ARCTIC Acute Rapid Cooling of Traumatic Injuries of the CNAP Compound nerve action potential
Cord study CNS Central nervous system
ARDS Acute respiratory distress syndrome CNT-​2 Concentrative nucleoside transporter type 2
ASA American Society of Anesthesiologists COMT Catechol-O-methyl transferase
ASA PS American Society of Anesthesiologists COX Cycloxygenase
Physical Status COX-​2 Cyclooxygenase-​2
ASIA American Spinal Injury Association CPB Cardiopulmonary bypass
ASICs Acid-​sensing ion channels CPP Cerebral perfusion pressure
ATP Adenosine triphosphate CPR Cardiopulmonary resuscitation
AV Atrioventricular CRP C-​reactive protein
AVM Arteriovenous malformations CRPS Chronic regional pain syndrome
Aβ Amyloid-​beta CSF Cerebrospinal fluid
BAC Balloon-​assisted coiling CSWS Cerebral salt wasting syndrome
xi

xii a bbreviations

CT Computed tomography HF Heart failure


CTA CT angiography HHT Hereditary Haemorrhagic Telangiectasia
CTP CT-​perfusion HIF-​1α Hypoxia-​inducible factor 1 alpha
Cv Venous concentration HS Hypertonic saline
CVA Cerebrovascular accident Hz Hertz
CVR Cerebrovascular resistance IADL Instrumental activities of daily living
DA1 Dopamine type 1 IARS International Anesthesia Research Society
DA2 Dopamine type 2 IBA1 Ionized calcium binding adaptor molecule 1
DBH dopamine β-​hydroxylase IBV Intracranial blood volume
DBS Deep brain stimulation/​stimulator ICA Internal carotid artery
DCI Delayed cerebral ischaemia ICH Intracranial haemorrhage
DDAVP desmopressin acetate ICP Intracranial pressure
DIND Delayed ischemic neurological deficit ICU Intensive care unit
DL Direct laryngoscopy ICV Intracranial volume
DLPFC Dorsolateral prefrontal cortex IHAST Intraoperative Hypothermia for Aneurysm
DMN Default Mode Network Surgery Trial
DOAC Direct acting oral anticoagulant IIT Intensive insulin therapy
DOPA Dihydroxyphenylalanine IL-​6 Interleukin-​6
DpMe Deep mesencephalic reticular formation IOM Intra-operative neurophysiological monitoring
DR Dorsal raphe ION Ischemic optic neuropathy
DRG Dorsal root ganglion IOM Intra-operative neurophysiological monitoring
DVT Deep vein thrombosis IONM Intraoperative neurophysiological monitoring
DWI diffusion-​weighted imaging IPG Internal pulse generator
ECG Electrocardiogram IPL Inferior parietal lobule
ECMO Extracorporeal membrane oxygenation IQ Intelligence quotient
ECoG Electrocorticography IV-​tPA Intravenous tissue-​type plasminogen activator
ECT Electroconvulsive therapy K Potassium
ED Effective dose K2P Two-​pore-​domain potassium channel
EEG Electroencephalography Kv Voltage-​gated potassium channel
EG Endothelial glycocalyx LA Local anaesthesia
EMG Electromyography LAT-​1 Large neutral amino-​acid transporter type 1
ENS Enteric nervous system LC Locus coeruleus
EP Evoked potentials LD Lumbar drain/​drainage
ESL Endothelial surface layer LDF Laser Doppler flowmetry
ESO European Stroke Organization LDT Laterodorsal tegmentum
ET Endotracheal tube LGICs Ligand-​gated ion channels
ETCO2 End-​tidal carbon dioxide LH Luteinizing hormone
ETV Endoscopic third ventriculostomy LMA Laryngeal mask airway
EVD External ventricular drain/​drainage LMWH Low molecular weight heparin
FDA Food and Drug Administration LoRR Loss of righting reflex
FFP Fresh frozen plasma LOX Lipoxygenase
FiO2 Fraction of inspired oxygen LP Lactate:pyruvate
FLAIR Fluid-​attenuated inversion recovery LVH Left ventricular hypertrophy
fMRI Functional magnetic resonance imaging MABL Maximal allowable blood loss
FOUR Full outline of unresponsiveness MAC Minimum alveolar concentration
FSH Follicle stimulating hormone MAC Monitored anaesthesia care
FV Flow velocity MADRS Montgomery-​Asberg Depression Rating Scale
GA General anaesthesia MAO Monoamine oxidase
GABA Gamma-​aminobutyric acid MAO-​B Monoamine oxidase-​B
GCS Glasgow Coma Scale MAOIs MAO inhibitors
GH Growth hormone MAP Mean arterial blood pressure
GI Gastrointestinal MCA Middle cerebral artery
GLUT-​1 Glucose transporter type 1 MCI Mild cognitive impairment
GPi Globus pallidus internus MCT-​1 Monocarboxylic acid transport type 1
H reflex Hoffmann’s reflex MDD Major depressive disorder
Hb Haemoglobin MDR-​1 Multidrug resistance gene
HCN Hyperpolarization-​activated cyclic MEG Magnetoencephalography
nucleotide-​gated MEN-​1 Multiple endocrine neoplasia type 1
HD Hydrocephalus MEP Motor evoked potentials
xi

 abbreviations xiii

MER Microelectrode recordings PICC Peripherally inserted central catheter


MERCI Mechanical Embolus Removal in Cerebral PIN Pressure inside the endoscope
Ischemia trial PION Posterior ischemic optic neuropathy
MH Malignant hyperthermia PIV Pressure-​induced vasodilation
miRNA Micro-​RNA PKA Protein kinase A
ml Millilitres PKC Protein kinase C
MLS Manual-​in-​line stabilization PNMT Phenylethanolamine N-​methyl transferase
MnPO Median preoptic nucleus PnO Pontine reticular nucleus, oral part
MOCAIP Morphological clustering and analysis of ICP pulse PNS Parasympathetic nervous system
mPFC Medial prefrontal cortex POCD Postoperative cognitive dysfunction
mps Metres per second PONV Postoperative nausea and vomiting
MRI Magnetic resonance imaging PORC Postoperative residual curarization/​Postoperative
mRNA Messenger RNA residual neuromuscular block
mRS Modified Rankin score POVL Postoperative vision loss
Mx Mean flow velocity reactivity PPT Pedunculopontine tegmentum
N2O Nitrous oxide PPV Positive prediction value
nAChR Nicotinic acetylcholine receptor PRES Posterior reversible encephalopathy syndrome
NANC non-​adrenergic non-​cholinergic neurotransmitter PRx Pressure reactivity index
Nav Voltage-​gated sodium PSI Patient state index
NCF Nucleus cuneiformis PtiO2 Brain tissue oxygenation
NGF Nerve growth factor PZ Parafacial zone
NICU Neurological intensive care unit RA Rheumatoid arthritis
NIHSS National Institutes of Health Stroke Scale RBC Red blood cell
NIRS Near infrared spectroscopy RCRI Revised cardiac risk index
NMB Neuromuscular block RCT Randomized controlled trial
NMDA N-​methyl-​D-​aspartate RE Response entropy
NMS Neuroleptic malignant syndrome REM Rapid eye movement
NO Nitric oxide/​Nitrogen monoxide RLN Recurrent laryngeal nerve
NOS Nitric oxide synthase RN Raphe nuclei
NPH Normal pressure hydrocephalus RNA Ribonucleic acid
NPPB Normal perfusion pressure breakthrough ROI Region of interest
NPY Neuropeptide Y ROS Reactive oxygen/​oxidative species
NREM Non-​REM Rout Resistance to CSF outflow
NS Nociceptive specific RSI Rapid sequence induction
NSAIDs Non-​steroidal anti-​inflammatory drugs rSO2 Regional cerebral oxygenation
NSF N-​ethyl maleimide sensitive factor rTPA Recombinant tissue plasminogen activator
NSM Neurogenic stunned myocardial R-​type High-​voltage-​activated calcium channels
NSQIP National Surgical Quality Improvement Program Risk RVM Rostroventralmedial medulla
OPP Ocular perfusion pressure RVP Rapid ventricular pacing
OR Operating room SA Sinoatrial
ORx Near-​infrared spectroscopy SAH Subarachnoid haemorrhage
OSA Obstructive sleep apnoea SBP Systolic blood pressure
OWLS Oral and written language scale SBT Spontaneous breathing test
PaCO2 Partial pressure of arterial carbon dioxide SCI Spinal cord injury
PACU Post-​anaesthesia care unit SE State entropy
PAG Periaqueductal grey SE Status epilepticus
PaO2 Partial pressure of arterial oxygen SEP Sensory evoked potentials
PB Parabrachial nucleus SI Primary somatosensory cortex
PCA Posterior cerebral artery SIADH Syndrome of inappropriate antidiuretic hormone
PCA Patient-​controlled analgesia secretion
PCC Prothrombin complex concentrate sICH Symptomatic intracerebral haemorrhage
PC-​MRI Phase-​contrast MRI SII Secondary somatosensory cortex
PCOM Posterior communicating SjvO2 Supra normal jugular venous oxygen saturation
PD Parkinson’s disease SMA Supplemental motor area
PEEP Positive end-​expiratory pressure SMT Spinomesencephalic tract
PET Positron emission tomography SNACC Society for Neuroscience in Anesthesiology and
PFC Prefrontal cortex Critical Care
PFO Patent foramen ovale SNAPs Synaptosomal-​associated protein
PGE2 Prostaglandin E2 SNARE Soluble NSF receptor
xvi

xiv a bbreviations

SNS Sympathetic nervous system TNF-​α Tumour necrosis factor α


SPECT Single-​photon emission CT tPA Tissue plasminogen activator
SRT Spinoreticular tract TRP Transient receptor potential
SSEP Somatosensory evoked potentials TRPM TRP melastatin receptor
SSRIs Selective serotonin and norepinephrine reuptake TRPV TRP vanilloid receptor
inhibitors TSH Thyroid stimulating hormone
STAIR Stroke Therapy Academic Industry Roundtable VAE Venous air embolism
STN Subthalamic nucleus VEP Visual Evoked Potentials
STT Spinothalamic tract VIP Vasoactive intestinal protein
SVS Slit ventricle syndrome VLPO Ventrolateral preoptic nucleus
SWS Slow-​wave sleep vPAG Ventral periaqueductal gray
TBI Traumatic brain injury VPL Ventroposterolateral
TCA Tricyclic antidepressant VPS Ventriculoperitoneal shunt
TCD Transcranial Doppler sonography VR-​1 Vanilloid receptor
TCS Transcranial stimulation VRL-​1 Vanilloid-​like receptor 1
TDF Thermal diffusion flowmetry VTA Ventral tegmental area
TEE Transoesophageal echocardiogram WDR Wide dynamic range
THx High temporal resolution WFNS World Federation of Neurological Surgeons
THx Therapeutic hypothermia ZO Zona occludens
TIVA Total intravenous anaesthetic β-​ARK β-​adrenergic receptor
TMN Tuberomamillary nucleus
xv

Contributors

Corey Amlong, Department of Anesthesiology, University of Gerald Glick†, Department of Medicine, Rush Medical
Wisconsin School of Medicine and Public Health, USA College, USA

Michael Avidan, Department of Anesthesiology, Washington David B. Glick, Department of Anesthesia & Critical Care,
University School of Medicine, USA University of Chicago, USA

Federico Bilotta, Department of Anesthesiology, Critical Care and Kerstin Göbel, Department of Neurology, University Hospital
Pain Medicine, Sapienza University of Rome, Italy Münster, Germany

Stefan Bittner, Department of Neurology, Johannes Gutenberg Shaun E. Gruenbaum, Department of Anesthesiology, Yale
University Mainz, Germany University School of Medicine, USA

Manfred Blobner, Klinik für Anaesthesiologie der Technischen Richard E. Harris, Department of Anesthesiology, University of
Universität München, Klinikum rechts der Isar, Germany Michigan Medical School, USA

Ansgar Brambrink, Department of Anesthesiology, Columbia Laura B. Hemmer, Department of Anesthesiology and
University, USA Neurological Surgery, Northwestern University, Feinberg School
of Medicine, USA
Douglas A. Colquhoun, Department of Anesthesiology, University
of Michigan Medical School, USA Eric J. Heyer, Departments of Anesthesiology and Neurology,
Columbia University, USA
Michael Crimmins, Walter Reed National Military Medical Center,
Department of Neurology, Neurosurgery and Critical Care, USA Ulrike Hoffmann, Department of Anesthesiology, Duke
University, USA
Zeyd Ebrahim, Department of General Anesthesiology,
Anesthesiology Institute, Cleveland Clinic, USA Paola Hurtado, Anesthesiology Department, Hospital Clìnic de
Barcelona, Spain.
Margaret K. Menzel Ellis, Portland VA Medical Center,
Assistant Professor of Anesthesiology, Oregon Health & Science Adam K. Jacob, Department of Anesthesiology and Perioperative
University, USA Medicine, Mayo Clinic College of Medicine, USA

Kristin Engelhard, Department of Anesthesiology, University M. Luke James, Departments of Anesthesiology and Neurology,
Medical Center of the Johannes Gutenberg-​University Mainz, Duke University, USA
Germany
Max B. Kelz, Department of Anesthesiology and Critical Care,
Neus Fàbregas, Anesthesiology Department, Hospital Clìnic de University of Pennsylvania Perelman School of Medicine, USA
Barcelona, Spain
Klaus Ulrich Klein, Department of Anesthesia, General Intensive
Ehab Farag, Department of General Anesthesia and Outcomes Care and Pain Management, Medical University of Vienna,
Research, Anesthesiology Institute, Cleveland Clinic, USA Austria

Heidrun Lewald, Klinik für Anaesthesiologie der Technischen Ines P. Koerner, Department of Anesthesiology & Perioperative
Universität München, Klinikum rechts der Isar, Germany Medicine, Department of Neurological Surgery, Oregon Health &
Science University, USA
Katherine M. Gelber, Department of Anesthesiology, Cedars-​Sinai
Medical Center, USA
xvi

xvi c ontributors

Antoun Koht, Department of Anesthesiology, Neurological Jamie Sleigh, Department of Anaesthesia and Pain Medicine,
Surgery, and Neurology, Northwestern University, Feinberg School Waikato Clinical Campus, University of Auckland, New Zealand
of Medicine, USA
Tod B. Sloan, Department of Anesthesia, University of Colorado
Sandra L. Kopp, Department of Anesthesiology and Perioperative School of Medicine, USA
Medicine, Mayo Clinic College of Medicine, USA
Martin Smith, Department of Neuroanaesthesia and Neurocritical
Brian P. Lemkuil, Department of Anesthesiology, University of Care, The National Hospital for Neurology and Neurosurgery,
California San Diego, USA University College London Hospitals, UK

Pirjo Manninen, Department of Anesthesia, Toronto Sulpicio G. Soriano, Department of Anesthesiology, Perioperative
Western Hospital University Health Network, University of and Pain Medicine, Boston Children's Hospital, Harvard Medical
Toronto, Canada School, USA

Nathan Manning, Departments of Neurosurgery and Christiane G. Stäuble, Klinik für Anaesthesiologie der
Radiology, Columbia University Medical Centre, New York Technischen Universität München, Klinikum rechts der Isar,
Presbyterian, USA Germany

Ross P. Martini, Department of Anesthesiology and Perioperative Harald Stefanits, Department of Neurosurgery, Medical
Medicine, Oregon Health & Science University, USA University of Vienna, Austria

Craig D. McClain, Department of Anesthesiology, Perioperative Erica J. Stein, Department of Anesthesiology, The Ohio State
and Pain Medicine, Boston Children's Hospital, Harvard Medical University, USA
School, USA
Magnus Teig, Department of Anesthesiology, University of
Andrew McKinstry-​Wu, Department of Anesthesiology and Michigan Medical School, USA
Critical Care, University of Pennsylvania, USA
J. Richard Toleikis, Department of Anesthesiology, Rush
Sven G. Meuth, Department of Neurology, Institute of University School of Medicine, USA
Translational Neurology, Westfälische-​Wilhelms University
Münster, Germany Timur M. Urakov, Department of Neurosurgery, University of
Miami, Jackson Memorial Hospital, USA
Philip M. Meyers, Departments of Radiology and Neurological
Surgery, Columbia University, USA Lashmi Venkatraghavan, Department of Anesthesia, Toronto
Western Hospital, University of Toronto, Canada
Edward C. Nemergut, Department of Anesthesiology, University
of Virginia Health System, USA Phillip E. Vlisides, Department of Anesthesiology, University of
Michigan Medical School, USA
Adam D. Niesen, Department of Anesthesiology and Perioperative
Medicine, Mayo Clinic College of Medicine, USA Laszlo Vutskits, Department of Anesthesiology, Pharmacology
and Intensive Care, University Hospitals of Geneva, Department
Jeffrey J. Pasternak, Department of Anesthesiology and of Basic Neuroscience, University of Geneva Medical School,
Perioperative Medicine, Mayo Clinic College of Medicine, USA Switzerland

Piyush Patel, VA Medical Center, University of California San Michael Y. Wang, University of Miami, Miller School of
Diego, USA Medicine, USA

Adrian Pichurko, Department of Anesthesiology, Northwestern Tasha L. Welch, Department of Anesthesiology and Perioperative
University, Feinberg School of Medicine, USA Medicine, Mayo Clinic College of Medicine, USA

Andrea Reinprecht, Department of Neurosurgery, Medical David R. Wright, Departments of Anesthesiology & Pain Medicine
University of Vienna, Austria and Neurological Surgery, University of Washington, USA

Robert D. Sanders, Department of Anesthesiology, University of Zhongcong Xie, Department of Anesthesia, Critical Care and
Wisconsin, USA Pain Medicine, Massachusetts General Hospital, Harvard Medical
School, USA
Anne Sebastiani, Department of Anesthesiology, University
Medical Center of the Johannes Gutenberg University Mainz, Sophia C. Yi, Department of Anesthesiology, University of
Germany California San Diego, USA

Deepak Sharma, Department of Anesthesiology & Pain Medicine,


University of WashingtonUSA
xvi

Digital media
accompanying the book

Individual purchasers of this book are entitled to free personal ac- The corresponding media can be found on Oxford Medicine
cess to accompanying digital media in the online edition. Please Online at: www.oxfordmedicine.com/otneuroanesthesiology
refer to the access token card for instructions on token redemption If you are interested in access to the complete online edition,
and access. please consult with your librarian.
These online ancillary materials, where available, are noted with
iconography throughout the book.
Q Cases and multiple-​choice questions
xvi
1

SECTION 1

Neuroscience
in Anaesthetic Practice

1 Neural Mechanisms of Anaesthetics 3


Andrew McKinstry-​Wu and Max B. Kelz
2 Intracranial Pressure 17
Harald Stefanits, Andrea Reinprecht, and Klaus Ulrich Klein
3 Cerebral Physiology 27
Stefan Bittner, Kerstin Göbel, and Sven G. Meuth
4 Introduction to Electroencephalography 35
Michael Avidan and Jamie Sleigh
5 The Autonomic Nervous System 47
David B. Glick, Gerald Glick†, and Erica J. Stein
6 Neuromuscular Junction: Anatomy and Physiology,
Paralytics, and Reversal Agents 61
Christiane G. Stäuble, Heidrun Lewald, and Manfred Blobner
7 Principles of Neuroprotection 77
Sophia C. Yi, Brian P. Lemkuil, and Piyush Patel
8 Neurotoxicity of General Anaesthetics 93
Margaret K. Menzel Ellis and Ansgar Brambrink
9 Neurobiology of Acute and Chronic Pain 111
Adrian Pichurko and Richard E. Harris
2
3

CHAPTER 1

Neural Mechanisms
of Anaesthetics
Andrew McKinstry-​Wu and Max B. Kelz

Introduction Ligand-​Gated Ion Channels


The first public demonstration of general anaesthesia took place Ligand-​gated ion channels (LGICs) are common targets for vola-
in 1846. Over 170 years later, a majority of the estimated 234 mil- tile, gaseous, and potent intravenous agents. They provide an easily
lion annual surgical procedures worldwide are performed under understood mechanism for modulating individual neural activity
general anaesthesia (1). Nevertheless, general anaesthetics remain and offer a plausible method for altering large-​scale neural effects.
poorly understood as a unique class of drug that has infallible General anaesthetics variously affect multiple LGICs, the two most
clinical efficacy with a narrow therapeutic window. Despite their common being potentiation of inhibitory anionic channels and in-
pervasive use, there is a lack of basic knowledge of where and hibition of excitatory cationic channels. In fact, the vast majority
how anaesthetics produce both their desirable and unintended of general anaesthetics demonstrate specific actions at one or both
side effects. of two LGICs: potentiation of the anionic gamma-​aminobutyric
Apparent similarities in dose-​dependent behavioural effects acid (GABA)-​gated GABAA receptor, and inhibition of the cationic
among gaseous, volatile, and intravenous general anaesthetics glutamate-​and-​glycine-​gated N-​methyl-​D-​aspartate (NMDA)
led to the historical belief that all general anaesthetics shared receptor.
a single molecular mechanism of action. Older theories of an- Inhibitory Ligand-​Gated Ion Channel Potentiation
aesthetic action relied on common chemical properties of the GABA is the most common inhibitory neurotransmitter in the
anaesthetics to explain their common effects, such as the asso- central nervous system (CNS.) The GABAA-​receptor, an abundant
ciation of lipid solubility with anaesthetic potency (the Meyer-​ GABA effector site, is a heteropentameric ligand-​gated chlorine-​
Overton rule). Ultimately, these early theories fell out of favour selective ion channel responsible for GABA’s inhibitory effects in the
with the realization that anaesthetics could exert their actions CNS. Importantly, it is also a crucial functional target of most po-
in lipid-​free protein preparations. Subsequently, many molecular tent intravenous agents and volatile anaesthetics (2–​7). Volatile and
targets of individual anaesthetics have been identified. With the intravenous anaesthetics that affect the GABAA-​receptor enhance
discovery of each new molecular target, the fallacy of a unitary endogenous GABAergic signalling at pharmacologically relevant
molecular mechanism of anaesthesia becomes more apparent. concentrations, and at higher concentrations can directly open the
The past twenty years have demonstrated that knowledge of both channel (5–​10). Synaptic potentiation of GABAA-​receptors affects
the specific molecular targets, as well as their location in dis- size and duration of rapid, phasic, inhibitory postsynaptic potentials.
crete neural circuits, is a prerequisite to any real understanding Potentiation at extrasynaptic receptors, in contrast, alters baseline
of anaesthetic hypnosis. Hence, the molecular, neuronal, cir- membrane potential through tonic chloride currents (11). The net
cuit, and network targets of anaesthetics are all critical to our effect of these actions is to decrease the chance that the postsynaptic
neuroscientific framework of how these agents produce revers- neuron will fire an action potential in the presence of pharmacologic-
ible unconsciousness. ally relevant concentrations of many general anaesthetics. Mounting
evidence suggests that it is the extrasynaptic, tonic inhibition that is
the primary method through which general anaesthetics produce
Molecular Mechanisms of Anaesthetic their effects (12). Specific mutations in GABAA-​receptor subunits at
Hypnosis known anaesthetic binding sites produce attenuation to anaesthetic
The breadth of molecular targets of general anaesthetics highlights effects of specific agents, both in vitro and in vivo. Mutations in the
the diversity of molecular mechanisms sufficient to produce anaes- alpha subunit of the GABAA-​receptor reduce the effect of volatile
thetic hypnosis. Inhaled and intravenous anaesthetics act on di- anaesthetics and benzodiazepines, while beta subunit mutations
verse protein targets to exert their hypnotic effects: ion channels, attenuate the effects of intravenous and volatile anaesthetics (3, 5).
G-​protein coupled receptors, and constituents of the electron trans- This suggests a critical role for action at the GABAA-​receptor in pro-
port chain, among others (Figure 1.1). ducing on-​target anaesthetic effects.
4

4 Section 1 neuroscience in anaesthetic practice

Mt
KV1 HCN K2P NaV NMDA Glycine GABA mAch nAch TTCa RTCa Complex
I

Ethers

Halothane

Propofol

Etomidate

Barbiturates

Ketamine

Nitrous
Oxide/
Xenon

Dexmedeto-
midine

Figure 1.1 Summary of the effect of anaesthetic drugs on molecular targets relevant to anaesthetic hypnosis.
Light blue circles represent activation or potentiation, dark blue circles indicate inhibition, and white circles indicate no effect. Circles with more than one colour are
present where different agents of a single anaesthetic class have differing effects. Where interactions have not been explored in the literature, no circle is present.
Kv1.1: Shaker-​related voltage-​gated potassium channel HCN: Hyperpolarization-​activated cyclic nucleotide-​gated channel, K2P: Two-​pore potassium channels, NMDA: N-​
methyl D-​aspartate receptor, Glycine: Glycine receptor, GABA: gamma-​aminobutyric acid receptor, mAch: muscarinic acetylcholine receptor, nAch: nicotinic acetylcholine
receptor, TTCa: T-​type calcium channel, RTCa: R-​type calcium channel, Mt complex I: Complex I of the electron transport chain (NADH: ubiquinone oxidoreductase).

Glycine receptors are the other significant inhibitory, anionic Excitatory Ligand-​Gated Ion Channel Inhibition
LGICs in the CNS. This receptor family is found mostly in the Many general anaesthetics act to inhibit excitatory LGICs, a
brainstem and spinal cord. Like GABAA-​receptors, glycine re- complementary effect to their potentiation of inhibitory LGICs.
ceptors are heteropentameric chlorine channels, and are directly Glutamate is the primary excitatory neurotransmitter of the CNS.
activated or potentiated by volatile and many intravenous anaes- Among glutamate’s targets is the NMDA receptor (where it has gly-
thetics (13–​16). Evidence for the functional importance of gly- cine as a co-​receptor). NMDA receptors are the functional target for
cine receptors to anaesthetic action is not as robust as that for the a significant number of general anaesthetics. Like the extrasynapic
GABAA-​receptor. Glycine receptor mutations can produce diver- GABA receptors responsible for tonic currents, NMDA receptors
gent responses to anaesthetic endpoints. Site-​specific mutations do not produce the fast postsynaptic transmission responsible
of the glycine receptor that alter in vitro receptor sensitivity to for excitatory postsynaptic potentials, but acts presynaptically,
volatile and potent intravenous anaesthetics do not always pro- postsynaptically, and extrasynaptically, and can affect synaptic plas-
duce associated changes in immobility or hypnotic sensitivity in ticity (21). All known noncompetitive NMDA antagonists severely
vivo (6, 17). Specifically with propofol, the glycine receptor may disturb consciousness, with many acting as general anaesthetics at
not contribute to immobility. A structural analogue of propofol sufficient concentrations (22). The gas anaesthetics nitrous oxide
that potentiates glycine (but not GABA) receptor signalling, 2,6 and xenon, as well as the intravenous agent ketamine, all act pri-
di-​tert-​butylphenol, lacks any immobilizing effects in vivo (7, marily as NMDA receptor antagonists, while many of the volatile
18, 19). Similarly, a Q266I point mutation introduced into the anaesthetics possess NMDA antagonist activity in addition to their
α1 subunit of the glycine receptor that decreases receptor sen- effects on other putative anaesthetic targets (23–​26).
sitivity to isoflurane unexpectedly conferred hypersensitivity to The nicotinic acetylcholine receptor, a ligand-​gated nonspecific
the immobilizing properties of both isoflurane and enflurane in cation channel, is inhibited by volatile anaesthetics at clinically
mice. These results suggest that glycine receptors containing the relevant concentrations. While that inhibition does not mediate
α1 subunit are unlikely to mediate immobilizing properties of anaesthetic hypnosis, it may mediate amnesia and analgesic ef-
anaesthetics (20). fects of volatile anaesthetics (27–​30). Moreover, central cholinergic
5

Chapter 1 neural mechanisms of anaesthetics 5

signalling via nicotinic receptors appears important for anaesthetic historically inhibition had only been seen at higher concentrations
emergence. Although blockade of cholinergic signalling may not (42). The role of sodium channels in volatile anaesthetic hypnosis
be sufficient to alter loss-​of-​righting-​reflex or Minimum alveolar is demonstrated by hypersensitivity to isoflurane and sevoflurane
concentration (MAC) concentration, it can still be sufficient to re- in mice with reduced activity in one voltage-​gated sodium channel
tard emergence from anaesthetic hypnosis (discussed later in this subtype, NaV1.6 (43).
chapter). Presynaptic voltage-​gated calcium channels are critical for neuro-
transmitter release and inhibited by general anaesthetics, making
Constitutively Active and Voltage-​Gated Ion Channels them likely anaesthetic targets. Low-​voltage-​activated T-​type
Members of the two-​pore-​domain potassium channel family calcium channels, which modulate cellular excitability through
(K2P) produce a continuous non-​inactivatable current that modi- regulating burst firing and pacemaker activity, are inhibited by
fies resting membrane potential and thus affects neuronal excit- clinically relevant concentrations of volatile and intravenous anaes-
ability (31). Volatile and gaseous anaesthetics directly activate thetics (44–​46). In vivo knockouts of T-​type calcium channels do
members of this family, including TREK-​1, TREK-​2, TASK-​1, not show changes in anaesthetic sensitivity to the loss of righting
TASK-​2, and TASK-​3. Anaesthetic activation of these K2P chan- reflex (LoRR), a traditional rodent equivalent endpoint to loss of
nels causes an increase in potassium efflux out of the cell leading to consciousness in humans, though they do have altered speed of in-
hyperpolarization. However, not every member of the K2P family duction and reaction to noxious stimuli (46, 47). This suggests that
is activated by anaesthetic exposure. Several members are insensi- the effects of anaesthetics upon these channels modulate the anaes-
tive to anaesthetics, while THIK-​1, TWIK-​2, TALK-​1, and TALK-​2 thetized state, rather than cause it. High-​voltage-​activated calcium
are actually closed by anaesthetic exposure. Mutations of two-​pore-​ channels (R-​type) are also sensitive to inhibition by volatile anaes-
domain potassium channels can abrogate sensitivity to activation thetics, and contribute to rhythmicity of thalamocortical circuits. R-​
by volatile and gaseous anaesthetics. Distinct gene mutations alter type knockouts display less electroencephalographic suppression at
volatile sensitivity versus sensitivity to gaseous anaesthetics (32). 1% isoflurane than their wild-​type counterparts. This suggests that
An in vivo knockout of one K2P, TREK-​1, caused an impressive 40% thalamic calcium channels are involved in isoflurane-​induced thal-
resistance to halothane and more modest resistance to other inhaled amic suppression, thought to contribute to unconsciousness (48).
anaesthetics, while leaving barbiturate sensitivity unchanged (33).
Hyperpolarization-​activated cyclic nucleotide-​ gated (HCN) G-​Protein-​Coupled Receptors
channels are tetrameric, relatively nonspecific cation channels that G-​protein-​coupled receptors make up the largest and most di-
activate with cell hyperpolarization (as opposed to depolarization.) verse family of membrane receptors. They comprise 4% of the en-
The Ih current, produced by HCN activation, is involved in pro- tire coding human genome and are the target for over a quarter
ducing long-​term potentiation, dendritic integration, control of of all current pharmaceuticals (49). Drugs that affect this receptor
working memory, and thalamocortical oscillations (34). Of the four superfamily are an integral part of anaesthetic practice, including
HCN isoforms, HCN1 is both abundant in the CNS and inhibited such diverse classes as opioids, vasopressors, and anticholinergics.
by volatile and intravenous agents. Agents as diverse as isoflurane, So, while these receptors are known targets for producing analgesia
ketamine, and propofol inhibit HCN1 at clinically relevant doses. and amnesia, there is little direct evidence that volatile-​anaesthetic-​
In in vivo models, this HCN1 inhibition plays a direct role in the induced hypnosis is primarily mediated via this superfamily of
hypnotic potency of these agents (35–​38). There is even debate that receptors. This is despite the fact that volatile anaesthetics do select-
NMDA receptor antagonists producing hypnosis do so not through ively activate G-​protein-​coupled receptors at pharmacologically-​
actions at the NMDA receptor itself, but through their inhibition relevant concentrations (50, 51). Ketamine very specifically
of HCN1 (37). The involvement of HCN channels in critical CNS interacts with a subset of olfactory receptors, which are a subgroup
processes and their inhibition by diverse anaesthetic agents suggest of G-​protein-​coupled receptors, though it is unclear if these inter-
a significant role for this channel in anaesthetic-​induced hypnosis. actions are in any way related to its anaesthetic actions (52).
Voltage-​gated potassium channels of the Kv1 family are recently
identified targets of volatile anaesthetics that contribute to suppres- Electron Transport Chain
sion of arousal. Flies with mutations in a gene coding for a member Unlike the previously discussed membrane-​bound proteins in the
of the Kv1.2 family (shaker) exhibit altered sensitivity to volatile an- cell’s outer membrane, components of complex I, a multi-​subunit
aesthetics, requiring higher doses than wild-​type controls to cease member of the respiratory chain, are putative anaesthetic targets lo-
movement (39). Sevoflurane enhances currents in members of the cated in the inner mitochondrial membrane. Animal models with
Kv1 family, with other clinically used volatiles also affecting Kv1 mutations in specific subunits of complex I, GAS-​1 in C. elegans
currents, suppressing firing in the central medial thalamus (40). and Ndufs4 in mice, are hypersensitive to volatile anaesthetics. This
Kv1 channel inhibitors infused into the central medial thalamus hypersensitivity phenotype is strictly mirrored across evolution up
are able to reverse continuous low-​dose sevoflurane anaesthesia in to and including humans with complex I mutations (53–​55). The
animal models, as are antibodies against Kv channels (41). anaesthetic hypersensitivity phenotype is not present in all com-
Voltage-​gated sodium channels are a requisite for normal exci- plex I gene mutations, nor is it present with other electron transport
tatory neuronal function, as they are key to initiating and propa- chain mutations, indicating a specific interaction between volatile
gating action potentials. Their inhibition by volatile anaesthetics anaesthetics and precise complex I subunits. While halogenated
presynaptically results in a decreased likelihood of action poten- ethers and alkanes inhibit complex I function though interaction
tial propagation and decreased presynaptic neurotransmitter re- with the distal portion of the complex, volatile anaesthetics do not
lease. Several sodium channel subtypes are inhibited by volatile appear to disproportionately decrease ATP production in complex
anaesthetics in pharmacologically relevant concentrations, though I mutants. This dissociation suggests volatile anaesthetic hypnotic
6

6 Section 1 neuroscience in anaesthetic practice

action is not solely a result of disproportionate mitochondrial ener- anaesthetic depth can also distinguish wakefulness from sleep (59–​
getic disruption in those mutants, begging the persistent question 65). During anaesthesia and sleep, thalamic nuclei and wake-​active
of how these mutations cause anaesthetic hypersensitivity (56–​58). nuclei, collectively known as the reticular activating system, are
similarly inhibited (46, 66–​70). Parallels between the states extend
Systems Neuroscience and Anaesthetic to their functional effects—​in some cases anaesthesia can substi-
tute for sleep. Sleep debt does not accrue during prolonged periods
Effects: Discrete Nuclei and Local Networks of propofol-​induced unconsciousness, while propofol hypnosis ap-
There is incontrovertible evidence for anaesthetic drugs interacting pears to relieve previously incurred sleep debt (71–​73). Conversely,
with multiple molecular targets to affect behaviour, but anaesthetic sleep deprivation or administration of endogenous somnogens
hypnosis is impossible to explain by mere examination of molecular reduce the dose of anaesthetic required for hypnosis. In a parallel
binding events in isolation. The imperfect connection between vein, induction and maintenance of anaesthesia itself alters levels
molecular action and larger-​scale brain phenomena must be in- of endogenous somnogens (72, 74). Together, these data support
terpreted in light of relevant neuroanatomy. Complexities are intro- the theory that anaesthetic-​induced hypnosis stems in part from
duced by circuit-​level interactions—​neuronal hyperpolarization actions of anaesthetics on the neural circuits involved with en-
that reduces firing of a presynaptic inhibitory input can increase dogenous sleep-​wake control.
activity for the postsynaptic neuron resulting in a net increase of
circuit output. Evaluating the net contribution of anaesthetics on Arousal-​Promoting Nuclei of the Reticular
discrete brain regions to hypnosis provides a way to simplify the Activating System
massive complexity encountered at the molecular and neuronal The ascending reticular activating system, extending rostrally from
level without ignoring the fundamental circuitry of the CNS. the mid pons to the hypothalamus, basal forebrain, and thalamus,
was first identified more than half a century ago. Stimulation of the
Sleep and Arousal Pathways brain stem reticular formation causes cortical arousal during an-
General anaesthetics alter the activity of endogenous arousal cir- aesthetic states (75). Subsequently, discrete interacting neuronal
cuits. Such actions directly contribute to their hypnotic effects populations were found to be the arousal-​promoting components
(Figure 1.2). Anaesthetic-​induced unconsciousness is a non-​ of the activating system, including cholinergic, histaminergic, ad-
arousable behavioural state that shares many commonalities with renergic, serotonergic, dopaminergic, and orexinergic centres.
slow-​wave sleep (SWS) There is a functional loss in cortical con-
nectivity during both NREM sleep and anaesthetic hypnosis. Laterodorsal Tegmentum (LDT) and Pedunculopontine
Moreover, over much of the anaesthetic dose response range, the Tegmentum (PPT)
cortical electroencephalography (EEG) exhibits striking simi- These nuclei comprise two major cholinergic populations in the
larities, such that processed EEG measures developed to assess brainstem with the ability to regulate arousal state and promote

Frontal cortex
Mesial parietal cortex
Precuneus
Posterior cingulate cortex
Thalamus
Hippocampus
Mesopontine tegmental area

Amygdala
Ox
TMN
(HA) VTA
(DA)

DpMe
(Glut)
BF
(Ach/Glut/GABA) PPT/LDT
RN
(ACh)
(5-HT)
POA
(GABA/Gal)
vPAG
LC (Ne)/PB
(DA)
(Glut)
PZ
(GABA) Pno

Figure 1.2 Cortical and subcortical (inset) structures affected by anaesthetic agents and potentially contributing to hypnosis.
Arrows indicate the ascending reticular activating system, both the anterior branch passing through the basal forebrain before ascending to the cortex and the posterior
branch extending into the cortex via the thalamus. The primary neurotransmitters associated with their respective subcortical structures are listed in parentheses.
BF: basal forebrain, Ox: orexin field, TMN: tuberomamillary nucleus, VTA: ventral tegmental area, DpMe: deep mesencephalic reticular formation, PPT/​
LDT: pedunculopontine tegmentum/​laterodorsal tegmentum, LC/​PB: locus coeruleus/​parabrachial nucleus, PnO: pontine oralis, PZ: parafacial nucleus, vPAG: ventral
periaqueductal grey, RN: raphe nucleus, POA: preoptic area. HA: histamine, DA: dopamine, Glut: glutamate, Ach: acetylcholine, GABA: gamma-​aminobutyric acid,
5-​HT: serotonin, Gal: galanin.
7

Chapter 1 neural mechanisms of anaesthetics 7

wakefulness or REM sleep. These cholinergic neurons densely systemic effects (92, 93). Clearly, a more complex local microcir-
innervate the midline and intralaminar thalamic nuclei and thal- cuitry awaits discovery.
amic reticular nucleus and alter thalamic activity from bursting to
Deep Mesencephalic Reticular Formation (DpMe)
spiking (76). Direct effects of these nuclei on anaesthetic-​induced
hypnosis are unknown. Despite this, the PPT is in a region that is Over the decades, many studies have demonstrated that electrical
associated with pain-​induced movement, the inactivation of which stimulation in the DpMe reliably induces cortical activation in an-
leads to a significant decrease of isoflurane MAC (77). aesthetized animals. These presumptively glutamatergic neurons
project to the thalamus, hypothalamus, and basal forebrain, where
Locus Coeruleus (LC) they increase their firing rates prior to the onset of wakefulness,
The Locus Coeruleus is the site of the brain’s largest collection of and fire more slowly during SWS (94). These glutamatergic neurons
noradrenergic neurons. As with many of the other monoaminergic are possibly part of a previously poorly recognized arm of the as-
systems, the LC diffusely innervates the brain by projecting directly cending arousal system that potentially includes the parabrachial
to the cortex, thalamus, hypothalamus, basal forebrain, amygdala, nucleus as well.
hippocampus, and other subcortical systems. State-​dependent
modulation of activity within the LC has long been proposed as Hypocretin/​Orexinergic Neurons
an essential means of regulating arousal. Changes in the activity The orexin signalling system exerts potent wake-​promoting and
of LC neurons occur before, and are predictive of, changes in an wake-​stabilizing effects, and plays an important role in modulating
organism’s behavioural state (78). Through its actions on alpha1 and anaesthetic emergence. As with the monoaminergic wake-​active
beta receptors, firing of LC neurons promotes wakefulness through systems, the orexin system displays state-​dependent firing pat-
actions on the medial septum, the medial preoptic area, and the terns with maximal activity during active wakefulness and silence
substantia innominata within the basal forebrain. Activity in the during SWS (95). Anatomically, these neurons project to all of the
LC modulates thalamocortical circuits, switching the tone of thal- monoaminergic groups along with extending to the basal forebrain,
amocortical neurons from the burst pattern of slow-​wave sleep to midline thalamic nuclei, and other regions known to participate in
a spiking pattern that characterizes wakefulness. Consequently, the regulation of arousal. When signalling of these neurons is im-
optogenetically driven LC activity causes transitions from SWS to paired, narcolepsy with cataplexy ensues (96). Local application of
wakefulness (79). Under deep isoflurane anaesthesia, artificially orexin excites target neurons expressing either of the two orexin Gq-​
driven LC activity has been shown to cause EEG desynchron- coupled neurotransmitter receptors, including the LDT, LC, RN,
ization. Similarly, artificially induced firing of the LC speeds emer- basal forebrain (BF), and thalamocortical neurons. Halogenated
gence from isoflurane anaesthesia (80). However, the LC is not the ethers, propofol, and pentobarbital inhibit orexinergic neuronal
sole source of adrenergically driven arousal. Noradrenergic popu- activity, and genetic knockout of these neurons results in delayed
lations outside of the LC, such as the A1 and A2 brainstem groups, emergence from isoflurane and sevoflurane anaesthesia without
also may contribute to the regulation of sleep, wakefulness, and an- affecting sensitivity to anaesthetic induction (97–​99). In the case
aesthesia (81–​83). of barbiturate anaesthesia, the pharmacologic inverse is true as
well: intracerebroventricular injection of orexin speeds emergence,
Pontine Reticular Nucleus, Oral Part (PnO) and orexin1-​receptor blockade negates this effect (100). The case
Neurons in this large region (which includes the sublaterodorsal of delayed emergence without altered induction in orexinergic de-
nucleus) receive cholinergic, orexinergic, and GABAergic inputs ficient animals highlights the intriguing possibility that distinct
and include wakefulness-​promoting and REM-​on populations. populations of neurons may unilaterally and differentially impact
PnO activity also modifies anaesthetic action. GABAergic activity the process of entering into or exiting from the anaesthetic state.
at the PnO produces resistance to induction without significant
effects on emergence (84–​87). Electrical stimulation at the PnO Wake-​Promoting Neurons of the Basal Forebrain
causes an increase in functional connectivity under continuous The BF encompasses heterogeneous populations of neurons ac-
isoflurane anaesthesia, similarly suggesting anaesthetic antag- tive in arousal and sleep that modify anaesthetic state and sensi-
onist actions (88). This region highlights the critical importance tivity. GABA agonists microinjected at the BF potentiate systemic
of neuroanatomic and neurochemical compartments: unlike most intravenous and volatile anaesthetic effect and duration, as do
other regions of brain, increased GABA levels in the PnO pro- electrolytic lesions of the medial septum within the BF (101, 102).
mote wakefulness. Other seemingly paradoxical effects occur in The BF sits atop the ventral extrathalamic relay and receives inte-
this region: wakefulness is actually impaired by local delivery of grated arousal inputs from caudal structures. Within the BF, there
adenosine or acetylcholine into PnO, or alternatively, promoted are wake-​active cholinergic neurons, wake-​active glutamatergic
by local delivery of orexin or GABA (89–​91). Cholinergic input to neurons, wake-​ a ctive parvalbumin-​ c ontaining GABAergic
the PnO originates from the LDT and PPT, while the orexinergic neurons, and sleep-​active somatostatin-​containing GABAergic
input arises from the hypothalamus. Divergent responses to adeno- neurons (103). The cholinergic neurons receive afferents from the
sine and GABA suggest that simple disinhibition of a single popu- LC, DpMe, Tuberomamillary nucleus (TMN), orexinergic neurons,
lation of PnO neurons is not sufficient to understand the actions parabrachial neurons, and glutamatergic neurons of the BF, and
of these neuromodulators. Further clouding the picture, micro- send widespread efferent projections to the cortex and hippo-
injections of pentobarbital within a region that has been termed campus, as well as back to the hypothalamus. Selective lesion of
the mesopontine tegmental area, which overlaps with a significant cholinergic neurons within the nucleus basalis of the BF prolongs
portion of the PnO and some neighbouring structures, have been behavioural effects of propofol and pentobarbital (104). Increased
shown to induce hypnosis similar to systemic administration of a cholinergic activity of the basal forebrain during wakefulness is re-
larger general anaesthetic dose, while nearby injections lack any sponsible for the fluctuations in cortical acetylcholine levels. The
8

8 Section 1 neuroscience in anaesthetic practice

cholinergic neurons stimulate cortical activation, and the resultant CNS serotonin. The median raphe contributes little to producing
increased discharge frequency, along with increased activity of or modulating anaesthetic actions. In contrast, when the DR is si-
cortically projecting parvalbumin-​containing GABAergic neurons, lenced by calcium blockage or by lesion, sensitivity is increased to
underlies the desynchronized EEG that typifies wakefulness as well pentobarbital or halothane and cyclopropane, respectively (124,
as REM sleep (103, 105). 125). The raphe nuclei (RN) do display state-​dependent firing
similar to noradrenergic or histaminergic centres. Single unit re-
Parabrachial Nucleus
cordings, however, demonstrate that firing rates of serotonergic
Neurons in the parabrachial nuclei belong to a recently identi- neurons do not anticipate spontaneous changes in arousal state
fied glutamatergic arm of the anterior branch of the ascending (126). This suggests that activity of the DR is not causally linked to
arousal system and can themselves modify the anaesthetic state. changes in behavioural state.
Lesion of the parabrachial nuclei induces coma, while the sleep-​ Within the vPAG, there is a population of dopaminergic neurons
active parafacial zone (see Sleep-​active Neurons of the Basal that are wake-​active. This is in contrast to dopaminergic neurons of
Forebrain and Parafacial Zone) promotes SWS through inhibition the VTA or substantia nigra, which do not display state-​dependent
of glutamatergic parabrachial neurons that, in turn, project to the activity. However, the vPAG dopaminergic neurons appear not to
BF (106–​109). Electrical stimulation of the parabrachial nucleus is contribute to anaesthetic hypnosis. Instead, they modulate anal-
able to antagonize continuous low-​dose isoflurane administration gesia, with lesions of these neurons causing decreased reaction to
(110). This observed direct effect on anaesthetic sensitivity, as well noxious stimuli under general anaesthesia (127, 128).
as the parabrachial’s known anatomic and functional connection
with sleep-​active nuclei and other CNS regions that affect anaes- Sleep-​Active Neurons and Nuclei
thetic sensitivity and emergence, suggests a significant role for the
When compared to the large numbers of known arousal-​
parabrachial nucleus in maintaining or emerging from an anaes-
promoting, wake-​active nuclei, there are few neural populations
thetic state. However, this has yet to be fully explored.
that are predominantly active during SWS or REM sleep with de-
Ventral Tegmental Area (VTA) creased activity during wakeful states. Neurons with sleep-​active
A midbrain dopaminergic and GABAergic region associated with firing patterns are most commonly found in the preoptic area and
both arousal and reward, the VTA receives diverse inputs and has BF (though populations do exist elsewhere, including in the pons
widespread outputs, including the prefrontal cortex, cingulate and cortex.) These populations act as a network counterbalance to
gyrus, hippocampus, amygdala, and nucleus acumbens. Reflecting many of the arousal-​promoting populations discussed previously.
those widespread connections, it is functionally diverse, with roles Just as there are many examples of anaesthetics depressing those
in motivation, cognition, and arousal (111). Dopaminergic neurons arousal centres, there is evidence for sleep-​active neurons being
of the VTA do not appear to change their firing rate across sleep–​ directly and indirectly activated by certain anaesthetics.
wake. Consequently, VTA dopamine neurons have not been linked Preoptic Area: Ventrolateral Preoptic Nucleus (VLPO) and
to modulation of spontaneous arousal. Nevertheless, dopamin- Median Preoptic Nucleus (MnPO)
ergic neurons are suppressed by GABA, and other neurons within
In the earliest attempts to identify specific populations of sleep-​
the VTA have circadian-​dependent activity (112–​116). Artificially
promoting neurons, a definitive role was assigned to the preoptic
driving dopamine release from the VTA, administration of methyl-
anterior hypothalamus. This broad region encompasses the VLPO,
phenidate, or more selective pharmacologic agents that act on D1-​
which lies within the anterior hypothalamus on the ventral floor
like receptors, will all accelerate anaesthetic emergence, and can
at the level of the optic chiasm, as well as the MnPO, which strad-
even reverse anaesthesia produced by continuous administrations
dles the decussation of the anterior commissure. Neurons with
of propofol or isoflurane (117–​120).
heightened activity during SWS are found throughout the preoptic
Tuberomamillary Nucleus (TMN) area, with the greatest concentration found in VLPO. Neurons of
The sole source for histamine in the CNS, the histaminergic neurons the VLPO are more active during SWS and REM sleep. Changes
of the TMN have widespread projection throughout the brain and in VLPO activity precede the changes in an organism’s behavioural
have activity tightly correlated with arousal state (121). Though state. Retrograde and anterograde labelling studies show the VLPO
centrally administered histamine causes potent arousal, and central is reciprocally interconnected with many wake-​promoting nuclei,
H1 antagonists produce sedation, the role of the TMN in modifying including the histaminergic TMN, serotonergic RN, noradrenergic
general anaesthesia is circumscribed. Lesions of the TMN increase LC, and the cholinergic LDT and PPT, as well as the orexinergic
sensitivity to isoflurane-​induced LoRR, but leave propofol, keta- neurons of the hypothalamus. VLPO neurons contain the inhibi-
mine, and barbiturate sensitivity unchanged (122). Cell-​specific tory neurotransmitters GABA and galanin and are thus ideally
knockout of GABAA receptors in histaminergic cells likewise positioned to coordinate and reciprocally inhibit wake-​active as-
produced no change in propofol hypnotic sensitivity. These phe- cending reticular activating nuclei.
nomena were observed despite the fact that histaminergic neurons The role of the preoptic area in promoting and modulating the
lacking GABAA receptors were resistant to hyperpolarization by anaesthetic state appears connected to its regulation of sleep and
propofol—​strong evidence that histaminergic neurons are not crit- wake. Destructive lesions of VLPO cause long-​lasting insomnia.
ical to induction or maintenance of propofol hypnosis (123). Bilateral lesions of the VLPO also cause a biphasic change in an-
aesthetic sensitivity that possibly relates to insomnia: at six days
Dorsal Raphe (DR) and Ventral Periaqueductal Gray (vPAG) post-​destruction, animals showed resistance to isoflurane LoRR,
The serotonergic neurons of the raphe nuclei have diffuse pro- while 24 days post-​lesion, subjects showed hypersensitivity to
jections throughout the brain and comprise the largest source of isoflurane LoRR. Such changes have been hypothesized to arise
9

Chapter 1 neural mechanisms of anaesthetics 9

due to accumulating sleep pressure as VLPO lesioned animals The central thalamus has long been known to be critical
fail to sleep (129, 130). Isoflurane directly depolarizes GABAergic to arousal, with small lesions of the area leading to gross dis-
neurons in VLPO. Acute resistance to isoflurane induction seen orders of consciousness (136). The anterior intralaminar nuclei,
following VLPO lesions suggests that in normal conditions, an- of which the central medial thalamus (CMT) is a part, receive
aesthetic activation of VLPO contributes to induction. Except for significant ascending cholinergic innervation from the BF and
ketamine, every other general anaesthetic appears to depolarize brainstem. Microinjection of nicotine into the CMT is suffi-
and recruit putative sleep-​active VLPO neurons. The population cient to reverse continuous systemic sevoflurane anaesthesia in
of neurons throughout the preoptic area (including the VLPO, animal models. This effect is replicated with an infusion of anti-
MnPO, and surrounding area) that are active during recovery SWS bodies against potassium channels in the Kv1 family (41, 137).
(after sleep deprivation) significantly overlap with the population In slice recordings of neurons of the CMT, sevoflurane reduces
of neurons activated with systemic administration of hypnotic firing, which is, in turn, reversed by administration of a Kv1 in-
doses of dexmedetomidine. This reinforces earlier findings sug- hibitor (40). Activity of the CMT appears to be critical for both
gesting dexmedetomidine acts on native preoptic sleep circuits to sleep and anaesthesia, as changes in high-​f requency oscillations
produce hypnosis (131, 132). that occur at both the onset of sleep and anaesthetic-​induced
Similar to the VLPO, MnPO sleep-​ a ctive neurons are loss of consciousness occur first in the CMT, rapidly followed
GABAergic and fire more rapidly several seconds prior to sleep by cortical changes (68). More generally, thalamic inactiva-
onset. Although not as broadly activated by anaesthetics as the tion as measured by reduced blood flow has been associated
VLPO, some inhaled anaesthetics also appear to activate putative with anaesthetic-​induced unconsciousness, although whether
sleep-​active MnPO neurons (133). Microinjection of benzodi- this is causal or secondary to unconsciousness itself remains
azepines, propofol, and pentobarbital into the MnPO has been unclear (138).
shown to induce SWS (72, 134, 135). Due to the state-​dependent
firing pattern of MnPO neurons, they have been assigned an im- Neocortex and Limbic Cortex
portant role in the initiation of sleep. The MnPO is known to send
The effects of anaesthetics are particularly heterogeneous across the
inhibitory projections to multiple arousal systems, including the
neocortex, in stark contrast to the largely antiquated ‘wet blanket’
orexinergic neurons in the hypothalamus, in a manner similar to
theory of anaesthetic action as a general neuronal depressant. The
the VLPO.
extent to which the primary sensory cortices are affected by many
Sleep-​active Neurons of the Basal Forebrain and Parafacial general anaesthetics remains unclear. These primary processing
Zone (PZ) areas are still able to maintain normal or near-​normal responses
While arousal-​promoting neurons of the BF and pons are well es- to evoked potentials while longer-​latency potentials are inhibited.
tablished and their effects on anaesthetic states detailed, it is only In contrast, the function of higher-​order association cortices and
recently that sleep-​active groups in these structures were identi- portions of the entorhinal cortex are markedly impaired by general
fied. Although they interact with arousal centres known to influ- anaesthetics. The mesial parietal cortex, the anterior and posterior
ence anaesthetic actions, the effects of these newly detailed groups cingulate cortices, and the precuneus are deactivated in sleep and
on the anaesthetic state and the effects of anaesthetics on their ac- anaesthesia, as measured indirectly by changes in cerebral blood
tion remain to be investigated. Parvalbumin-​positive GABAergic flow (139–​141). Given the variability of anaesthetic effect on in-
neurons of the BF are wake-​active and strongly promote arousal, dividual cortical areas, hypnotic effects of anaesthetics may either
while another subset of GABAergic neurons expressing somato- prove to be a result of direct actions upon the cortex or could be a
statin in the BF comprise a sleep-​active group. Somatostatin posi- result of network-​level perturbations that include cortical and sub-
tive, GABAergic neurons of the BF exhibit specific increases in cortical interactions.
local unit activity during SWS. Optogenetic stimulation of these
neurons can induce SWS (103). The parafacial zone is a pontine re-
gion lateral to the seventh nerve containing SWS-​active GABAergic Brain Networks and Anaesthesia
neurons. Those sleep-​active, inhibitory neurons project directly The specific molecular target(s) of an anaesthetic may not yield
onto arousal-​promoting glutamatergic neurons of the parabrachial immediate insight into mechanisms by which the drug pro-
nucleus, which, in turn, project to arousal-​promoting neurons of duces unconsciousness. Complete understanding requires de-
the BF. Artificial stimulation of those PZ GABAergic neurons pro- tailed knowledge of drug effects on their molecular targets, as
duces SWS (108). Anaesthetic effects on the PZ and sleep-​active well as how ensuing changes in the activity of neurons and glia
neurons of the BF await further study. affect local and distant circuits in addition to the global impact
on networks. The relevant actions of anaesthetics, and hence their
Thalamus common mechanism, may thus be their ultimate disruption of
The thalamus is a critical structure that plays at least three major network function-​decreasing functional information integration.
roles in wakefulness and consciousness. First, in terms of levels of Understanding discrete effects of anaesthetics on receptors, indi-
consciousness, the thalamus is an important conduit for arousal vidual neurons, and brain nuclei are necessary but not sufficient
pathways from the brainstem. Second, in terms of the content of in isolation. To investigate anaesthetic disruption of network level
consciousness, it is a major relay station for sensory information en processes, large-​scale brain activity studies employ functional
route to the cortex. Third, in terms of the organization of conscious magnetic resonance imaging (fMRI), positron emission tomog-
experience, the higher-​order nuclei of the thalamus play a critical raphy (PET), magnetoencephalography (MEG), or high-​density
role in facilitating corticocortical coherence and communication. electroencephalography (EEG).
01

10 Section 1 neuroscience in anaesthetic practice

Thalamocortical Network anaesthetic-​induced unconsciousness. The parietal lobe con-


Thalamocortical networks are key to information integration, as tains multiple primary sensory cortices. Executive function
they gate ascending sensory information, ascending arousal signals, depends upon the frontal cortex. Consequently, effective com-
and modulate corticocortical relays. Disruption of the system illus- munication between the two regions is considered necessary for
trates the importance of this network, which results in disorders consciousness. It follows that disruption of that communication
of consciousness (136, 142). Shifting from cortically dominant to would serve to produce anaesthetic-​induced unconsciousness
thalamus-​dominant thalamocortical circuits during anaesthetic-​ (152, 153). However, rather than a simple complete breakdown
induced loss of consciousness corroborates a model of propofol of all frontal-​parietal communication, anaesthetic-​induced un-
hypnosis whereby GABAergic activation results in strengthened consciousness is associated with a specific disruption of feed-
thalamocortical connections and an intrinsic thalamically-​driven back communication between the frontal and parietal cortices.
alpha (8–​12 Hz) electrical rhythm (143, 144). Evidence from Thus, although there is effective information transfer from the
animal models that changes in thalamic rhythms slightly precede parietal cortex forward to the frontal cortex (feed-​forward),
related cortical rhythms at the time of anaesthetic-​induced loss reciprocal information transfer from the frontal cortex to the
of consciousness also lends credence to the theory of thalamus-​ parietal (feedback) is impaired with the onset of anaesthetic-​
rhythm-​driven thalamocortical loop as critical to anaesthetic hyp- induced unconsciousness (154). This pattern of feedback in-
nosis (68). Other animal models using field potentials within the hibition has proven remarkably robust over anaesthetics with
thalamus and cortical EEG show that ketamine/​xylazine-​induced varying molecular mechanisms of actions, from ketamine, to
loss of consciousness coincides with a shift in the dominant dir- volatile agents, to propofol (155). That this pattern of feedback
ection of information transfer from cortex-​thalamus during con- or top-​down processing is also impaired in vegetative states sug-
sciousness to thalamus-​cortex (145). Counter to the argument of a gests that this could be a general process for unconsciousness
predominant thalamocortical disruption by anaesthetics, slice and (156, 157).
chronic in vivo recordings suggest that corticocortical connections
are preferentially disrupted by volatile anaesthetics, while thalamo- Global Distance Connectivity and Information
cortical connections remain intact (146), Integration Capacity
Evidence from functional connectivity studies of the thal- In addition to evidence that anaesthetics may exert their effects
amus and cortex, which rely on relative blood flow as measured through specific networks such as those mentioned previously,
via PET or fMRI, rather than the electrical measurements of EEG anaesthetic action on large-​scale brain network organization and
and subcortical electrodes, do not demonstrate tight association generalized ability for information integration may be the source
of thalamus and cortex. Instead, their functional connectivity is of their unconsciousness-​inducing effects. Analyses of whole-​brain
eliminated with the onset of anaesthetic-​induced unconsciousness, networks under general anaesthesia show increased local informa-
purportedly through a silencing of the thalamus (147). While this tion exchange but impaired longer distance communication (158).
loss of functional connectivity between the cortex and thalamus This change from global connectedness to a more local pattern oc-
was first described with isoflurane and halothane, a similar pattern curs for both volatile anaesthetics and propofol, and has been seen
was also described with dexmedetomidine-​induced loss of con- in with the distinct measurement modalities of both EEG and fMRI
sciousness (148). Recent studies have further solidified the pattern (159). While not all analyses have specifically found an increase
of reduced thalamocortical connectivity using both ketamine and of ‘small-​worldness’ of the networks (local over long-​distance),
sevoflurane, making it a robust finding across multiple anaesthetic decreases in network efficiency are common, and therefore the
classes (149, 150). amount of information that could be transmitted and integrated
The difference in conclusions between methods using rela- by a network is diminished in the presence of a general anaesthetic
tive blood flow as a proxy for neural activity and those directly (160–​163).
measuring electrical signatures of that activity may be a result of dif-
ferent temporal and spatial resolutions between the two strategies.
Simultaneous measures of EEG and fMRI during a slow propofol-​ Conclusions
induced loss of consciousness addressed this disparity, and dem- While primary anaesthetic pharmacologic effects necessarily
onstrated a distinct phenomenon: isolation of the thalamocortical occur at the molecular level, the diversity of both general an-
network from sensory input at the point of maximum slow-​wave aesthetics’ molecular targets and their subsequent cellular-​level
activity on EEG, and the emergence of a sensory-​responsive primi- actions does not clearly translate into obvious behavioural com-
tive cortical network independent of the isolated thalamocortical monalities observed in vivo. Only at the level of neural circuits
network (151). The functional compartmentalization through thal- can we begin to appreciate the common effects among the diverse
amocortical isolation corresponds with the other EEG evidence, array of individual anaesthetic drugs. Distinguishing the prox-
suggesting emergence of thalamically driven rhythms during imate cause and secondary effects culminating in anaesthetic hyp-
anaesthetic-​induced unconsciousness (143, 144). Only with func- nosis remains a significant challenge for the field. Identifying the
tional isolation would the dominant rhythm driver be thalamus ra- critical anaesthetic induced network adaptations in both cortical
ther than corticocortical connections. and subcortical circuits truly responsible for loss of consciousness
itself, as opposed to circuit level side effects of anaesthetic drug
Corticocortical Networks exposure, represents another significant challenge. Resolving
As sensory integration and processing are integral to awareness, these challenges will benefit both the field of anaesthesiology as
the disruption of these functions are a predicted hallmark for well as neuroscience.
1

Chapter 1 neural mechanisms of anaesthetics 11

Summary 6. Mascia M, Trudell JR, Harris AR. Specific binding sites for alcohols and
anesthetics on ligand-​gated ion channels. Proceedings of the National
◆ Anaesthetics act upon a variety of protein targets to exert their Academy of Sciences. 2000;97(16):9305–​10.
hypnotic effects. 7. Hemmings HC, Akabas MH, Goldstein PA. Emerging molecular
mechanisms of general anesthetic action. Trends in Pharmacological
◆ Relevant hypnotic molecular targets of general anaesthetics in- Sciences. 2005;26(10):503–​10.
clude ligand-​gated ion channels (examples: GABA and NMDA 8. Jenkins A, Greenblatt EP, Faulkner HJ, Bertaccini E, Light A, Lin
receptors), voltage-​gated ion channels (examples: Kv1 and A,, et al. Evidence for a common binding cavity for three general
Nav1.6), constitutively active ion channels (example: K2P), G-​ anesthetics within the GABAA receptor. The Journal of Neuroscience.
protein-​coupled receptors, and respiratory complex I. 2001;21(6):RC136.
9. Li, X, Pearce, RA. Effects of halothane on GABA(A) receptor
◆ Anaesthetics elicit circuit-​level effects not readily predicted by kinetics: evidence for slowed agonist unbinding. The Journal of
their effects on single neurons of a given circuit. Neuroscience. 2000;20(3):899–​907.
10. Bieda M, MacIver B. Major role for tonic GABAA conductances in
◆ Anaesthetics can affect endogenous neural systems regulating anesthetic suppression of intrinsic neuronal excitability. Journal of
sleep and arousal, and such actions may produce or potentiate Neurophysiology. 2004;92(3):1658–​67.
their hypnotic effects. 11. Farrant M, Nusser Z. Variations on an inhibitory theme: phasic and
tonic activation of GABAA receptors. Nature Reviews Neuroscience.
◆ The thalamus is part of the ascending arousal system, serves as a
2005;6(3):215–​29.
relay for sensory information, and facilitates corticocortical com- 12. Bonin R, Orser B. GABAA receptor subtypes underlying
munication. While thalamic inactivation does not invariably lead general anesthesia. Pharmacology Biochemistry and Behavior.
to loss of consciousness, artificial stimulation can reverse hyp- 2008;90(1):105–​12.
nosis during continuous anaesthetic administration, presumably 13. Mascia M, Machu T, Harris A. Enhancement of homomeric glycine
through the ascending arousal pathways. receptor function by longchain alcohols and anaesthetics. British
Journal of Pharmacology. 1996;119(7):1331–​6.
◆ Disruption of function in cortical association areas by anaes- 14. Harrison NL, Kugler JL, Jones MV, Greenblatt EP, Pritchett DB.
thetics corresponds with changes of global network properties Positive modulation of human gamma-​aminobutyric acid type A and
during hypnosis, where long-​range connections decline and glycine receptors by the inhalation anesthetic isoflurane. Molecular
the total capacity for information integration decreases. It is Pharmacology. 1993;44(3):628–​32.
not yet clear whether direct action at those association centres 15. Beckstead M, Phelan R, Trudell J, Bianchini M, Mihic J. Anesthetic and
ethanol effects on spontaneously opening glycine receptor channels.
directly underlies network adaptations or whether the primary
Journal of Neurochemistry. 2002;82(6):1343–​51.
change occurs elsewhere and propagates to impair cortical 16. Yamakura T, Mihic SJ, Harris RA. Amino acid volume and
activity. hydropathy of a transmembrane site determine glycine and anesthetic
◆ Both thalamocortical networks as well as corticocortical patterns sensitivity of glycine receptors. The Journal of Biological Chemistry.
1999;274(33):23006–​12.
of connectivity are altered with anaesthetic hypnosis. Changes in
17. Quinlan JJ, Ferguson C, Jester K, Firestone LL, Homanics GE. Mice
both of these networks may reflect a final common marker or with glycine receptor subunit mutations are both sensitive and resistant
mediator of anaesthetic-​induced unconsciousness. to volatile anesthetics. Anesthesia and Analgesia. 2002;95(3):578–​82.
18. Ahrens J, Haeseler G, Leuwer M, Mohammadi B, Krampfl K, Dengler
Multiple-​Choice Questions R, et al. 2,6 di-​tert-​butylphenol, a nonanesthetic propofol analog,
modulates alpha1beta glycine receptor function in a manner distinct
Q Interactive multiple-​choice questions to test your knowledge from propofol. Anesthesia and Analgesia. 2004; 99(1):91–​6.
on this chapter can be found in the online appendix at www. 19. Krasowski MD, Jenkins A, Flood P, Kung AY. General anesthetic
oxfordmedicine.com/​otneuroanesthesiology. potencies of a series of propofol analogs correlate with potency
for potentiation of gamma-​aminobutyric acid (GABA) current at
the GABAA receptor but not with lipid solubility. The Journal of
References Pharmacology and Experimental Therapeutics. 2001; 297(1):338–​51.
1. Weiser TG, Regenbogen SE, Thompson KD, Haynes, AB, Lipsitz, 20. Borghese C, Xiong W, Oh S, Ho A, Mihic S, Zhang L, et al. Mutations
SR, Berry, WR, et al. An estimation of the global volume of M287L and Q266I in the glycine receptor α1 subunit change
surgery: a modelling strategy based on available data. Lancet. sensitivity to volatile anesthetics in oocytes and neurons, but not the
2008;372(9633):139–​44. minimal alveolar concentration in knockin mice. Anesthesiology.
2. Nelson, LE, Guo, TZ, Lu, J, Saper, CB, Franks, NP, Maze, M. The 2012;117(4):765–​71.
sedative component of anesthesia is mediated by GABAA receptors in 21. Mori H, Mishina M. Structure and function of the NMDA receptor
an endogenous sleep pathway. Nature Neuroscience. 2002;5(10):979–​84. channel. Neuropharmacology. 1995;34(10):1219–​37.
3. Jurd R, Arras M, Lambert S, Drexler B, Siegwart R, Crestani F, et al. 22. Flohr H, Glade U, Motzko D. The role of the NMDA synapse in general
General anesthetic actions in vivo strongly attenuated by a point anesthesia. Toxicology Letters. 1998;100–​101:23–​9.
mutation in the GABA(A) receptor beta3 subunit. The FASEB Journal. 23. Jevtović-​Todorović V, Todorović SM, Mennerick S, Powell S,
2003;17(2):250–​2. Dikranian K, Benshoff N, et al. Nitrous oxide (laughing gas) is an
4. Zecharia AY, Nelson LE, Gent TC, Schumacher M, Jurd R, NMDA antagonist, neuroprotectant and neurotoxin. Nature Medicine.
Rudolph U, et al. The involvement of hypothalamic sleep pathways 1998;4(4):460–​3.
in general anesthesia: Testing the hypothesis using the GABAA 24. Petrenko AB, Yamakura T, Fujiwara N, Askalany AR, Baba H,
receptor β3N265M knock-​in mouse. The Journal of Neuroscience. Sakimura K. Reduced sensitivity to ketamine and pentobarbital in mice
2009;29(7):2177–​87. lacking the N-​methyl-​D-​aspartate receptor GluRepsilon1 subunit.
5. Mihic J, Ye Q, Wick M, Koltchine V, Krasowski M, Finn S, et al. Sites Anesthesia and Analgesia. 2004;99(4):1136–​40.
of alcohol and volatile anaesthetic action on GABAA and glycine 25. Franks NP, Dickinson R, de Sousa SL, Hall AC, Lieb WR. How does
receptors. Nature. 1997;389(6649):385–​9. xenon produce anaesthesia? Nature. 1998;396(6709):324.
21

12 Section 1 neuroscience in anaesthetic practice

26. Armstrong S, Banks P, McKitrick T, Geldart C, Edge C, Babla R, et al. 46. Orestes P, Bojadzic D, Chow R, Todorovic S. Mechanisms and
Identification of two mutations (F758W and F758Y) in the N-​methyl-​ functional significance of inhibition of neuronal t-​type calcium
D-​aspartate receptor glycine-​binding site that selectively prevent channels by isoflurane. Molecular Pharmacology. 2009;75(3):542–​54.
competitive inhibition by xenon without affecting glycine binding. 47. Petrenko AB, Tsujita M, Kohno T, Sakimura K, Baba H. Mutation
Anesthesiology. 2012;117(1):38. of alpha1G T-​type calcium channels in mice does not change
27. Link J, Papadopoulos G, Dopjans D, Guggenmoos-​Holzmann I, anesthetic requirements for loss of the righting reflex and minimum
Eyrich K. Distinct central anticholinergic syndrome following general alveolar concentration but delays the onset of anesthetic induction.
anaesthesia. European Journal of Anaesthesiology. 1997;14(1):15–​23. Anesthesiology. 2007;106(6):1177–​85.
28. Meuret P, Backman S, Bonhomme V, Plourde G, Fiset P. Physostigmine 48. Joksovic P, Weiergräber M, Lee W, Struck H, Schneider T, Todorovic
reverses propofol-​induced unconsciousness and attenuation of S. Isoflurane-​sensitive presynaptic R-​type calcium channels contribute
the auditory steady state response and bispectral index in human to inhibitory synaptic transmission in the rat thalamus. The Journal of
volunteers. Anesthesiology. 2000;93(3):708. Neuroscience. 2009;29(5):1434–​45.
29. Flood P, Sonner J, Gong D, Coates K. Heteromeric nicotinic inhibition 49. Overington J, Al-​Lazikani B, Hopkins A. How many drug targets are
by isoflurane does not mediate MAC or loss of righting reflex. there? Nature Reviews Drug Discovery. 2006;5(12):993–​6.
Anesthesiology. 2002;97(4):902. 50. Ishizawa Y, Pidikiti R, Liebman P, Eckenhoff R. G protein-​coupled
30. Plourde G, Chartrand D, Fiset P, Font S, Backman SB. Antagonism receptors as direct targets of inhaled anesthetics. Molecular
of sevoflurane anaesthesia by physostigmine: effects on the auditory Pharmacology. 2002;61(5):945–​52.
steady-​state response and bispectral index. British Journal of 51. Peterlin Z, Ishizawa Y, Araneda R, Eckenhoff R, Firestein S. Selective
Anaesthesia. 2003;91(4):583–​6. activation of G-​protein coupled receptors by volatile anesthetics.
31. Millar J, Barratt L, Southan A, Page K, Fyffe R, Robertson B, et al. A Molecular and Cellular Neuroscience. 2005;30(4):506512.
functional role for the two-​pore domain potassium channel TASK-​1 52. Ho J, Perez-​Aguilar JM, Gao L, Saven JG, Matsunami H, Eckenhoff RG.
in cerebellar granule neurons. Proceedings of the National Academy of Molecular recognition of ketamine by a subset of olfactory G protein-​
Sciences. 2000;97(7):3614–​8. coupled receptors. Science Signaling. 2015;8(370):ra33.
32. Franks N. General anaesthesia: from molecular targets to neuronal 53. Kayser EB, Morgan PG, Sedensky MM. GAS-​1: a mitochondrial
pathways of sleep and arousal. Nature Reviews Neuroscience. protein controls sensitivity to volatile anesthetics in the nematode
2008;9(5):370–​86. Caenorhabditis elegans. Anesthesiology. 1999;90(2):545–​54.
33. Heurteaux C, Guy N, Laigle C, Blondeau N, Duprat F, Mazzuca M, 54. Quintana A, Morgan P, Kruse S, Palmiter R, Sedensky M. Altered
et al. TREK‐1, a K + channel involved in neuroprotection and general anesthetic sensitivity of mice lacking Ndufs4, a subunit of
anesthesia. The EMBO Journal. 2004;23(13):2684–​95. mitochondrial complex I. PloS ONE. 2012;7(8):e42904.
34. Wahl-​Schott C, Biel M. HCN channels: Structure, cellular regulation 55. Niezgoda J, Morgan P. Anesthetic considerations in patients with
and physiological function. Cellular and Molecular Life Sciences. mitochondrial defects. Pediatric Anesthesia. 2013;23(9):785–​93.
2008;66(3):470–​94. 56. Hanley P, Ray J, Brandt U, Daut J. Halothane, isoflurane and
35. Zhou C, Douglas J, Kumar N, Shu S, Bayliss D, Chen X. Forebrain sevoflurane inhibit NADH: Ubiquinone oxidoreductase (complex I) of
HCN1 channels contribute to hypnotic actions of ketamine. cardiac mitochondria. The Journal of Physiology. 2002;544(3):687–​93.
Anesthesiology. 2013;118(4):785. 57. Suthammarak W, Somerlot B, Opheim E, Sedensky M, Morgan P.
36. Zhou C, Liang P, Liu J, Ke B, Wang X, Li F, et al. HCN1 channels Novel interactions between mitochondrial superoxide dismutases and
Ccontribute to the effects of amnesia and hypnosis but not immobility the electron transport chain. Aging Cell. 2013;12(6):1132–​40.
of volatile anesthetics. Anesthesia and Analgesia. 2015;121(3):661–​6. 58. Manjeri GR, Rodenburg RJ, Blanchet L, Roelofs S, Nijtmans LG,
37. Chen X, Shu S, Bayliss DA. HCN1 channel subunits are a molecular Smeitink JA, et al. Increased mitochondrial ATP production capacity
substrate for hypnotic actions of ketamine. The Journal of Neuroscience. in brain of healthy mice and a mouse model of isolated complex
2009;29(3):600–​9. I deficiency after isoflurane anesthesia. Journal of Inherited Metabolic
38. Chen X, Shu S, Bayliss D. Suppression of Ih contributes to propofol-​ Disease. 2015;39(1):59–​65.
induced inhibition of mouse cortical pyramidal neurons. Journal of 59. Mashour GA. Consciousness unbound: toward a paradigm of general
Neurophysiology. 2005;94(6):3872–​83. anesthesia. Anesthesiology. 2004;100(2):428–​33.
39. Weber B, Schaper C, Bushey D, Rohlfs M, Steinfath M, Tononi G, et al. 60. John ER, Prichep LS, Kox W, Valdés-​Sosa P, Bosch-​Bayard J, Aubert E,
Increased volatile anesthetic requirement in short-​sleeping drosophila et al. Invariant reversible QEEG effects of anesthetics. Consciousness
mutants. Anesthesiology. 2009;110(2):313. and Cognition. 2001;10(2):165–​83.
40. Lioudyno M, Birch A, Tanaka B, Sokolov Y, Goldin A, Chandy 61. Pack CC, Berezovskii VK, Born RT. Dynamic properties of neurons in
G, et al. Shaker-​related potassium channels in the central medial cortical area MT in alert and anaesthetized macaque monkeys. Nature.
nucleus of the thalamus are important molecular targets for arousal 2001;414(6866):905–​8.
suppression by volatile general anesthetics. The Journal of Neuroscience. 62. Peltier SJ, Kerssens C, Hamann SB, Sebel PS, Byas-​Smith M, Hu X.
2013;33(41):16310–​22. Functional connectivity changes with concentration of sevoflurane
41. Alkire MT, Asher CD, Franciscus AM, Hahn EL. Thalamic anesthesia. NeuroReport. 2005;16(3):285–​8.
microinfusion of antibody to a voltage-​gated potassium channel 63. Massimini M, Ferrarelli F, Huber R, Esser SK, Singh H, Tononi G.
restores consciousness during anesthesia. Anesthesiology. Breakdown of cortical effective connectivity during sleep. Science.
2009;110(4):766–​73. 2005;309(5744):2228–​32.
42. Herold KF, Hemmings HC. Sodium channels as targets for volatile 64. Ferrarelli F, Massimini M, Sarasso S, Casali A, Riedner B, Angelini G,
anesthetics. Frontiers in Pharmacology. 2012;3:50. et al. Breakdown in cortical effective connectivity during midazolam-​
43. Pal D, Jones JM, Wisidagamage S, Meisler MH, Mashour GA. Reduced induced loss of consciousness. Proceedings of the National Academy of
Nav1.6 sodium channel activity in mice increases in vivo sensitivity to Sciences. 2010;107(6):2681–​6.
volatile anesthetics. PLoS ONE. 2015;10(8):e0134960. 65. Sleigh JW, Andrzejowski J, Steyn-​Ross A, Steyn-​Ross M. The bispectral
44. Study, RE. Isoflurane inhibits multiple voltage-​gated calcium index: A measure of depth of sleep? Anesthesia and Analgesia. 1999
currents in hippocampal pyramidal neurons. Anesthesiology. Mar 1;88(3):659–​61.
1994;81(1):104–​16. 66. Detsch O, Kochs E, Siemers M, Bromm B, Vahle-​Hinz C. Increased
45. Todorovic S, Perez-​Reyes E, Lingle C. Anticonvulsants but not general responsiveness of cortical neurons in contrast to thalamic neurons
anesthetics have differential blocking effects on different T-​type current during isoflurane-​induced EEG bursts in rats. Neuroscience Letters.
variants. Molecular Pharmacology. 2000;58(1):98–​108. 2002;317(1):9–​12.
31

Chapter 1 neural mechanisms of anaesthetics 13

67. Alkire MT, Haier RJ, Fallon JH. Toward a unified theory of phase of anesthesia and modulates hyperalgesia caused by sleep
narcosis: brain imaging evidence for a thalamocortical switch as deprivation. European Journal of Neuroscience. 2014;40(1):2264–​73.
the neurophysiologic basis of anesthetic-​induced unconsciousness. 88. Pillay S, Liu X, Baracskay P, Hudetz A. Brainstem stimulation
Consciousness and Cognition. 2000;9(3):370–​86. increases functional connectivity of basal forebrain-​paralimbic
68. Baker R, Gent T, Yang Q, Parker S, Vyssotski A, Wisden W, et al. network in isoflurane-​anesthetized rats. Brain Connectivity.
Altered activity in the central medial thalamus precedes changes in the 2014;4(7):523–​34.
neocortex during transitions into both sleep and propofol anesthesia. 89. Watson CJ, Soto-​Calderon H, Lydic R, Baghdoyan HA. Pontine
The Journal of Neuroscience. 2014;34(40):13326–​35. reticular formation (PnO) administration of hypocretin-​1 increases
69. Vahle-​Hinz C, Detsch O, Siemers M, Kochs E, Bromm B. Local GABAA PnO GABA levels and wakefulness. Sleep. 2008;31(4):453–​64.
receptor blockade reverses isoflurane’s suppressive effects on thalamic 90. Tanase D, Baghdoyan HA, Lydic R. Dialysis delivery of an adenosine
neurons in vivo. Anesthesia and Analgesia. 2001;92(6):1578–​84. A1 receptor agonist to the pontine reticular formation decreases
70. Steriade M. The corticothalamic system in sleep. Frontiers in Bioscience. acetylcholine release and increases anesthesia recovery time.
2003;8:d878–​99. Anesthesiology. 2003;98(4):912–​20.
71. Tung A, Bergmann B, Herrera S, Cao D, Mendelson W. Recovery from 91. Coleman C, Baghdoyan H, Lydic R. Dialysis delivery of an adenosine
sleep deprivation occurs during propofol anesthesia. Anesthesiology. A2A agonist into the pontine reticular formation of C57BL/​6J
2004;100(6):1419. mouse increases pontine acetylcholine release and sleep. Journal of
72. Tung A, Szafran M, Bluhm B, Mendelson W. Sleep deprivation Neurochemistry. 2006;96(6):1750–​9.
potentiates the onset and duration of loss of righting reflex induced by 92. Devor M, Zalkind V. Reversible analgesia, atonia, and loss
propofol and isoflurane. Anesthesiology. 2002;97(4):906. of consciousness on bilateral intracerebral microinjection of
73. Tung A, Lynch J, Mendelson W. Prolonged sedation with propofol in pentobarbital. Pain. 2001;94(1):101–​12.
the rat does not result in sleep deprivation. Anesthesia and Analgesia. 93. Abulafia R, Zalkind V, Devor M. Cerebral activity during the
2001;92(5):1232. anesthesia-​like state induced by mesopontine microinjection of
74. Kaputlu I, Sadan G, Ozdem S. Exogenous adenosine potentiates pentobarbital. The Journal of Neuroscience. 2009;29(21):7053–​64.
hypnosis induced by intravenous anaesthetics. Anaesthesia. 94. Datta S, MacLean R. Neurobiological mechanisms for the
1998;53(5):496–​500. regulation of mammalian sleep–​wake behavior: Reinterpretation
75. Moruzzi G, Magoun HW. Brain stem reticular formation and activation of historical evidence and inclusion of contemporary cellular
of the EEG. Electroencephalography and Clinical Neurophysiology. and molecular evidence. Neuroscience & Biobehavioral Reviews.
1949;1(4):455–​73. 2007;31(5):775–​824.
76. McCarley R. Neurobiology of REM and NREM sleep. Sleep Medicine. 95. Lee M, Hassani O, Jones B. Discharge of identified orexin/​hypocretin
2007;8(4):302–​30. neurons across the sleep-​waking cycle. The Journal of Neuroscience.
77. Jinks S, Bravo M, Satter O, Chan Y-​M. Brainstem regions affecting 2005;25(28):6716–​20.
minimum alveolar concentration and movement pattern during 96. Kilduff TS, Peyron C. The hypocretin/​orexin ligand–​receptor
isoflurane anesthesia. Anesthesiology. 2010;112(2):316. system: Implications for sleep and sleep disorders. Trends in
78. Berridge C, Schmeichel B, España R. Noradrenergic modulation of Neurosciences. 2000;23(8):359–​65.
wakefulness/​arousal. Sleep Medicine Reviews. 2012;16(2):187–​97. 97. Zecharia A, Franks N. General anesthesia and ascending arousal
79. Carter M, Yizhar O, Chikahisa S, Nguyen H, Adamantidis A, Nishino pathways. Anesthesiology. 2009;111(4):695.
S, et al. Tuning arousal with optogenetic modulation of locus coeruleus 98. Zhang L-​N, Li Z-​J, Tong L, Guo C, Niu J-​Y, Hou W-​G, et al. Orexin-​A
neurons. Nature Neuroscience. 2010;13(12):1526–​33. facilitates emergence from propofol anesthesia in the rat. Anesthesia
80. Vazey E, Aston-​Jones G. Designer receptor manipulations reveal and Analgesia. 2012;115(4):789.
a role of the locus coeruleus noradrenergic system in isoflurane 99. Kelz M, Sun Y, Chen J, Meng Q, Moore J, Veasey S, et al. An essential
general anesthesia. Proceedings of the National Academy of Sciences. role for orexins in emergence from general anesthesia. Proceedings of
2014;111(10):3859–​64. the National Academy of Sciences. 2008;105(4):1309–​14.
81. Kubota T, Anzawa N, Hirota K, Yoshida H, Kushikata T, Matsuki A. 100. Kushikata T, Hirota K, Yoshida H, Kudo M, Lambert DG, Smart D,
Effects of ketamine and pentobarbital on noradrenaline release from et al. Orexinergic neurons and barbiturate anesthesia. Neuroscience.
the medial prefrontal cortex in rats. Canadian Journal of Anaesthesia. 2003;121(4):855–​63.
1999;46(4):388–​92. 101. Ma J, Shen B, Stewart LS, Herrick IA, Leung LS. The
82. Yoshida H, Kushikata T, Kubota T, Hirota K, Ishihara H, Matsuki A. septohippocampal system participates in general anesthesia. The
Xenon inhalation increases norepinephrine release from the anterior Journal of Neuroscience. 2002; 22(2):RC200
and posterior hypothalamus in rats. Canadian Journal of Anaesthesia. 102. Leung S, Ma J, Shen B, Nachim I, Luo T. Medial septal lesion enhances
2001;48(7):651–​5. general anesthesia response. Experimental Neurology. 2013;247:419–​28.
83. Hirota K, Kushikata T. Central noradrenergic neurones and the 103. Xu M, Chung S, Zhang S, Zhong P, Ma C, Chang W-​C, et al. Basal
mechanism of general anaesthesia. British Journal of Anaesthesia. forebrain circuit for sleep-​wake control. Nature Neuroscience.
2002;87(6):811–​3. 2015;18(11):1641–​7.
84. Nelson AM, Battersby AS, Baghdoyan HA, Lydic R. 104. Leung S, Petropoulos S, Shen B, Luo T, Herrick I, Rajakumar, et al.
Opioid-​induced decreases in rat brain adenosine levels are Lesion of cholinergic neurons in nucleus basalis enhances response to
reversed by inhibiting adenosine deaminase. Anesthesiology. general anesthetics. Experimental Neurology. 2011;228(2):259–​69.
2010;111(6):1327–​33. 105. Lin S-​C, Brown R, Shuler M, Petersen C, Kepecs A. Optogenetic
85. Vanini G, Watson CJ, Lydic R, Baghdoyan HA. Gamma-​aminobutyric dissection of the basal forebrain neuromodulatory control of cortical
acid-​mediated neurotransmission in the pontine reticular formation activation, plasticity, and cognition. The Journal of Neuroscience.
modulates hypnosis, immobility, and breathing during isoflurane 2015;35(41):13896–​903.
anesthesia. Anesthesiology. 2009;109(6):978–​88. 106. Fuller P, Sherman D, Pedersen N, Saper C, Lu J. Reassessment of
86. Vanini G, Wathen B, Lydic R, Baghdoyan H. Endogenous GABA levels the structural basis of the ascending arousal system. The Journal of
in the pontine reticular formation are greater during wakefulness Comparative Neurology. 2011;519(5):933–​56.
than during rapid eye movement sleep. The Journal of Neuroscience. 107. Anaclet C, Lin J-​S, Vetrivelan R, Krenzer M, Vong L, Fuller P, et al.
2011;31(7):2649–​56. Identification and characterization of a sleep-​active cell group
87. Vanini G, Nemanis K, Baghdoyan H, Lydic R. GABAergic transmission in the rostral medullary brainstem. The Journal of Neuroscience.
in rat pontine reticular formation regulates the induction 2012;32(50):17970–​6.
41

14 Section 1 neuroscience in anaesthetic practice

108. Anaclet C, Ferrari L, Arrigoni E, Bass C, Saper C, Lu J, et al. The 129. Eikermann M, Vetrivelan R, Grosse-​Sundrup M, Henry M, Hoffmann
GABAergic parafacial zone is a medullary slow wave sleep-​promoting U, Yokota S, et al. The ventrolateral preoptic nucleus is not required
center. Nature Neuroscience. 2014;17(9):1217–​24. for isoflurane general anesthesia. Brain Research. 2011;1426:30–​7.
109. Kaur S, Pedersen N, Yokota S, Hur E, Fuller P, Lazarus M, et al. 130. Moore J, Chen J, Han B, Meng Q, Veasey S, Beck S, et al. Direct
Glutamatergic signaling from the parabrachial nucleus plays a activation of sleep-​promoting VLPO neurons by volatile anesthetics
critical role in hypercapnic arousal. The Journal of Neuroscience. contributes to anesthetic hypnosis. Current Biology. 2012;22(21).
2013;33(18):7627–​40. 131. Nelson LE, Lu J, Guo T, Saper CB, Franks NP, Maze M. The alpha2-​
110. Muindi F, Kenny J, Taylor N, Solt K, Wilson M, Brown E, et al. adrenoceptor agonist dexmedetomidine converges on an endogenous
Electrical stimulation of the parabrachial nucleus induces reanimation sleep-​promoting pathway to exert its sedative effects. Anesthesiology.
from isoflurane general anesthesia. Behavioural Brain Research. 2003;98(2):428–​36.
2016;306:20–​5. 132. Zhang Z, Ferretti V, Güntan İ, Moro A, Steinberg EA, Ye Z,
111. Beier KT, Steinberg EE, DeLoach KE, Xie S, Miyamichi K, Schwarz et al. Neuronal ensembles sufficient for recovery sleep and the
L, et al. Circuit architecture of VTA dopamine neurons revealed by sedative actions of α2 adrenergic agonists. Nature Neuroscience.
systematic input-​output mapping. Cell. 2015;162(3):622–​34. 2015;18(4):553–​61.
112. Yim CY, Mogenson GJ. Electrophysiological studies of neurons in the 133. Han B, McCarren HS, O’Neill D, Kelz MB. Distinctive recruitment
ventral tegmental area of Tsai. Brain Research. 1980;181(2):301–​13. of endogenous sleep-​promoting neurons by volatile anesthetics and a
113. Lee RS, Steffensen SC, Henriksen SJ. Discharge profiles of ventral nonimmobilizer. Anesthesiology. 2014;121(5):999–​1009.
tegmental area GABA neurons during movement, anesthesia, and the 134. Mendelson WB. Sleep induction by microinjection of
sleep-​wake cycle. The Journal of Neuroscience. 2001;21(5):1757–​66. pentobarbital into the medial preoptic area in rats. Life Sciences.
114. Luo AH, Aston-​Jones G. Circuit projection from suprachiasmatic 1996;59(22):1821–​8.
nucleus to ventral tegmental area: a novel circadian output pathway. 135. Tung A, Bluhm B, Mendelson WB. Sleep-​inducing effects of
European Journal of Neuroscience. 2009;29(4):748–​60. propofol microinjection into the medial preoptic area are blocked by
115. Luo AH, Georges FEE, Aston-​Jones GS. Novel neurons in ventral flumazenil. Brain Research. 2001;908(2):155–​60.
tegmental area fire selectively during the active phase of the diurnal 136. Schiff N. Central thalamic contributions to arousal regulation and
cycle. European Journal of Neuroscience. 2008;27(2):408–​22. neurological disorders of consciousness. Annals of the New York
116. Miller JD, Farber J, Gatz P, Roffwarg H, German DC. Activity of Academy of Sciences. 2008;1129(1):105–​18.
mesencephalic dopamine and non-​dopamine neurons across stages of 137. Alkire M, McReynolds J, Hahn E, Trivedi A. Thalamic microinjection
sleep and waking in the rat. Brain Research. 1983;273(1):133–​41. of nicotine reverses sevoflurane-​induced loss of righting reflex in the
117. Chemali J, Dort C, Brown E, Solt K. Active emergence from propofol rat. Anesthesiology. 2007;107(2):264.
general anesthesia is induced by methylphenidate. Anesthesiology. 138. Mhuircheartaigh R, Rosenorn-​Lanng D, Wise R, Jbabdi S, Rogers
2012;116(5):998. R, Tracey I. Cortical and subcortical connectivity changes during
118. Taylor N, Chemali J, Brown E, Solt K. Activation of D1 dopamine decreasing levels of consciousness in humans: A functional magnetic
receptors induces emergence from isoflurane general anesthesia. resonance imaging study using propofol. The Journal of Neuroscience.
Anesthesiology. 2013;118(1):30. 2010;30(27):9095–​102.
119. Solt K, Van Dort CJ, Chemali JJ, Taylor NE, Kenny JD, Brown 139. Maquet Cyclotron Research Centre. Functional neuroimaging of
EN. Electrical stimulation of the ventral tegmental area normal human sleep by positron emission tomography. Journal of
induces reanimation from general anesthesia. Anesthesiology. Sleep Research. 2000;9(3):207–​31.
2014;121(2):311–​9. 140. Kaisti KK, Långsjö JW, Aalto S, Oikonen V, Sipilä H, Teräs M, et al.
120. Solt K, Cotten J, Cimenser A, Wong K, Chemali J, Brown E. Effects of sevoflurane, propofol, and adjunct nitrous oxide on regional
Methylphenidate actively induces emergence from general anesthesia. cerebral blood flow, oxygen consumption, and blood volume in
Anesthesiology. 2011;115(4):791. humans. Anesthesiology. 2003;99(3):603–​13.
121. Takahashi K, Lin J-​S, Sakai K. Neuronal activity of histaminergic 141. Långsjö J, Alkire M, Kaskinoro K, Hayama H, Maksimow A, Kaisti
tuberomammillary neurons during wake–​sleep states in the mouse. K, et al. Returning from oblivion: Imaging the neural core of
The Journal of Neuroscience. 2006;26(40):10292–​8. consciousness. The Journal of Neuroscience. 2012;32(14):4935–​43.
122. Luo T, Leung SL. Involvement of tuberomamillary histaminergic 142. Schmid M, Singer W, Fries P. Thalamic coordination of cortical
neurons in isoflurane anesthesia. Anesthesiology. 2011;115(1):36–​43. communication. Neuron. 2012;75(4).
123. Zecharia A, Yu X, Götz T, Ye Z, Carr D, Wulff P, et al. GABAergic 143. Ching S, Cimenser A, Purdon PL, Brown EN, Kopell NJ.
inhibition of histaminergic neurons regulates active waking but not Thalamocortical model for a propofol-​induced alpha-​rhythm
the sleep–​wake switch or propofol-​induced loss of consciousness. The associated with loss of consciousness. Proceedings of the National
Journal of Neuroscience. 2012;32(38):13062–​75. Academy of Sciences. 2010;107(52):22665–​70.
124. Cui SY, Cui XY, Zhang J, Wang ZJ, Yu B. Diltiazem potentiates 144. Vijayan S, Ching S, Purdon PL, Brown EN, Kopell NJ.
pentobarbital-​induced hypnosis via 5-​HT 1A and 5-​HT 2A/​2C Thalamocortical mechanisms for the anteriorization of α rhythms
receptors: Role for dorsal raphe nucleus. Pharmacology, Biochemistry, during propofol-​induced unconsciousness. The Journal of
and Behavior. 2011; 99(4):556–​72. Neuroscience. 2013;33(27):11070–​5.
125. Roizen MF, White PF, Eger EI, Brownstein M. Effects of ablation 145. Kim S-​P, Hwang E, Kang J-​H, Kim S, Choi J. Changes in the
of serotonin or norepinephrine brain-​stem areas on halothane and thalamocortical connectivity during anesthesia-​induced transitions in
cyclopropane MACs in rats. Anesthesiology. 1978;49(4):252–​5. consciousness. NeuroReport. 2012;23(5):294.
126. Trulson ME, Jacobs BL. Raphe unit activity in freely moving 146. Raz A, Grady S, Krause B, Uhlrich D, Manning K, Banks M.
cats: correlation with level of behavioral arousal. Brain Research. Preferential effect of isoflurane on top-​down vs. bottom-​up pathways
1979;163(1):135–​50. in sensory cortex. Frontiers in Systems Neuroscience. 2014;8:191.
127. Lu J, Jhou T, Saper C. Identification of wake-​active dopaminergic 147. White N, Alkire M. Impaired thalamocortical connectivity in humans
neurons in the ventral periaqueductal gray matter. The Journal of during general-​anesthetic-​induced unconsciousness. NeuroImage.
Neuroscience. 2006;26(1):193–​202. 2003;19(2):402–​11.
128. Lu J, Nelson L, Franks N, Maze M, Chamberlin N, Saper C. Role of 148. Akeju O, Loggia ML, Catana C, Pavone KJ, Vazquez R, Rhee J, et al.
endogenous sleep‐wake and analgesic systems in anesthesia. Journal Disruption of thalamic functional connectivity is a neural correlate of
of Comparative Neurology. 2008;508(4):648–​62. dexmedetomidine-​induced unconsciousness. Elife. 2014;3:e04499.
51

Chapter 1 neural mechanisms of anaesthetics 15

149. Bonhomme V, Vanhaudenhuyse A, Demertzi A, Bruno M-​AA, Jaquet 156. Boly M, Garrido MI, Gosseries O, Bruno M-​AA, Boveroux P,
O, Bahri MA, et al. Resting-​state network-​specific breakdown of Schnakers C, et al. Preserved feedforward but impaired top-​down
functional connectivity during ketamine alteration of consciousness processes in the vegetative state. Science. 2011;332(6031):858–​62.
in volunteers. Anesthesiology. 2016;125(5):873–​88. 157. Thul A, Lechinger J, Donis J, Michitsch G, Pichler G, Kochs EF, et al.
150. Ranft A, Golkowski D, Kiel T, Riedl V, Kohl P, Rohrer G, et al. EEG entropy measures indicate decrease of cortical information
Neural correlates of sevoflurane-​induced unconsciousness identified processing in disorders of consciousness. Clinical Neurophysiology.
by simultaneous functional magnetic resonance imaging and 2016;127(2):1419–​27.
electroencephalography. Anesthesiology. 2016;125:861–​72. 158. Schröter M, Spoormaker V, Schorer A, Wohlschläger A, Czisch M,
151. Mhuircheartaigh R, Warnaby C, Rogers R, Jbabdi S, Tracey I. Kochs E, et al. Spatiotemporal reconfiguration of large-​scale brain
Slow-​wave activity saturation and thalamocortical isolation during functional networks during propofol-​induced loss of consciousness.
propofol anesthesia in humans. Science Translational Medicine. The Journal of Neuroscience. 2012;32(37):12832–​40.
2013;5(208):208ra148. 159. Li D, Voss LJ, Sleigh JW, Li X. Effects of volatile anesthetic agents
152. Imas OA, Ropella KM, Wood JD, Hudetz AG. Isoflurane on cerebral cortical synchronization in sheep. Anesthesiology.
disrupts anterio-​p osterior phase synchronization of flash-​ 2013;119(1):81–​8.
induced field potentials in the rat. Neuroscience Letters. 160. Liang Z, King J, Zhang N. Intrinsic organization of the anesthetized
2006;402(3):216–​2 1. brain. The Journal of Neuroscience. 2012;32(30):10183–​91.
153. Imas O, Ropella K, Ward B, Wood J, Hudetz A. Volatile 161. Lee H, Mashour G, Noh G-​J, Kim S, Lee U. Reconfiguration of
anesthetics disrupt frontal-​p osterior recurrent information network hub structure after propofol-​induced unconsciousness.
transfer at gamma frequencies in rat. Neuroscience Letters. Anesthesiology. 2013;119(6):1347.
2005;387(3):145–​50. 162. Monti M, Lutkenhoff E, Rubinov M, Boveroux P, Vanhaudenhuyse A,
154. Ku S-​W, Lee U, Noh G-​J, Jun I-​G, Mashour G. Preferential inhibition Gosseries O, et al. Dynamic change of global and local information
of frontal-​to-​parietal feedback connectivity is a neurophysiologic processing in propofol-​induced loss and recovery of consciousness.
correlate of general anesthesia in surgical patients. PloS ONE. PloS Computational Biology. 2013;9(10):e1003271.
2011;6(10). 163. Liu X, Ward D, Binder J, Li S-​J, Hudetz A. Scale-​free functional
155. Lee U, Ku S, Noh G, Baek S, Choi B, Mashour G. Disruption connectivity of the brain is maintained in anesthetized healthy
of frontal–​parietal communication by ketamine, propofol, and participants but not in patients with unresponsive wakefulness
sevoflurane. Anesthesiology. 2013;118(6):1264. syndrome. PLoS ONE. 2014;9(3).
61
71

CHAPTER 2

Intracranial Pressure
Harald Stefanits, Andrea Reinprecht,
and Klaus Ulrich Klein

Introduction and subarachnoidal spaces, rinsing metabolic compounds from the


central nervous system (CNS). It has been suggested that CSF has
This chapter on intracranial pressure (ICP) explains the following a ‘sink action’ by which metabolites produced in the brain during
topics: contents of the intracranial vault (brain parenchyma, cere- metabolic activity diffuse into CSF and thereby are removed from
brospinal fluid, arterial and venous blood), ICP waveforms, intra- the brain. Also, it has been suggested that CSF flow might be able
cranial elastance curve, intracranial hypertension, intracranial to cool the brain whenever it is needed. CSF plays an important
herniation syndromes, and monitoring of ICP. role in flushing cerebral metabolic toxins (e.g., beta amyloid) from
Alexander Monro (1733–​1817) first described ICP in 1783. By the interstitial fluid. This process is increased during natural sleep
describing that brain tissue is nearly incompressible and sur- by opening extracellular channels controlled by glial cells allowing
rounded by non-​expandable cranium, he suggested that intra- rapid influx of CSF into the brain.
cranial blood volume (IBV) remains constant. George Kellie Today, indications for monitoring of ICP include traumatic brain
(1720–​1779) stated that intracranial fluid could not be added or injury, intracerebral haemorrhage, subarachnoid haemorrhage
removed without simultaneous equivalent replacement or displace- (SAH), hydrocephalus, malignant infarction, oedema, infection,
ment. Francois Magendie (1783–​1855) first described the circula- and metabolic disorders (4). Measurement of ICP complements in-
tion of cerebrospinal fluid (CSF) flowing from the ventricles to the formation on cerebral perfusion pressure (CPP), cerebrovascular
spinal cord by discovering a small foramen in the roof of the fourth autoregulation, and compliance of the cerebrospinal system.
ventricle (foramen Magendie). In 1846, Sir George Burrows (1801–​
1887) constituted a reciprocal relationship between IBV and CSF.
The neurosurgeon Harvey Cushing (1869–​1939) and his co-​worker Contents of the Intracranial Vault (Brain
Lewis Weed endorsed the doctrine of Monro and Kellie by stating Parenchyma, CSF, Blood)
that with an intact cranium, the net sum of all intracranial vault
To understand the dynamics of ICP regulation, anatomical con-
volumes (brain tissue, blood, CSF volume) remains constant and
siderations have to be undertaken concerning the contents of the
that an increase in one component should cause a reduction in one
intracranial vault. The outlined anatomical landmarks are im-
or both of the other two components (1).
portant for the understanding of herniation syndromes and their
Investigation of CSF started in 1891, when Heinrich Quincke
clinical symptoms, since brainstem structures, cranial nerves, and
(1842–​1922) published his studies on lumbar puncture and the
arterial vessels are located close to the intracranial bottleneck areas
chemical investigation of CSF. In the early twentieth century, re-
and edges. The skull is the brain’s bony (stiff) hull that has only
petitive lumbar CSF puncture was widely used as first clinical
one big outlet for the medulla oblongata: the foramen magnum. It
method to determine ICP. Later in 1960, the neurosurgeon pioneer
contains all relevant compartments, including the brain tissue, CSF
Nils Lundberg published a thesis that largely influenced ICP moni-
spaces, and arterial and venous vessels (Figure 2.1 and Table 2.1).
toring (2), as measurements were first performed in the ventricles
over a period of several hours (3).
Numerous theories exist to explain why CSF surrounds the cere- Parenchyma
brum: a) buoyancy: the mass of the brain is about 1400 grams, The brain parenchyma including the cranial nerves makes up about
although the net weight of the brain suspended in the CSF is 85% of total intracranial volume (ICV). The largest part of the
equivalent to a mass of 25 grams. Therefore, the brain exists brain is the cerebrum (or telencephalon) with its two hemispheres,
in neutral buoyancy, allowing the brain to maintain its density separated by a vertical (sagittal) diaphragm, the falx cerebri. It is
without being impaired by its own weight. According to philoso- situated on top of the ‘core of the brain’, which is considered to be
pher Rudolf Steiner (1861–​1925), this exemption of the gravity the relay station for higher cognitive functions: the thalamus and
is essential for higher brain function; b) protection: CSF protects brainstem, parts of the diencephalon and mesencephalon, which
the brain to a certain extent against any impact; c) prevention of continues caudally to the pons and the medulla oblongata. Dorsally
brain ischaemia: CSF can be reduced to counteract brain oedema adjacent to the brainstem is the cerebellum, which is separated
when needed; d) chemical stability/​cooling/​clearing waste: CSF is from the cerebrum by an axial tent-​like diaphragm, the tentorium
produced in the ventricles and circulates through the ventricular cerebelli. The tentorial notch allows the caudal continuation of the
81

18 Section 1 neuroscience in anaesthetic practice

ultrafiltration from blood in the plexus choroideus at a rate of 250–​


500 ml/​24h. Its resorption sites are arachnoidal granulations close
to the venous sinuses and the spinal nerve roots. Production and re-
7
sorption are in a physiological balance. There are four major inner
CSF spaces, called the ventricles, which are connected to each other
1 and to the outer CSF space, i.e., the subarachnoid space. The lateral
ventricles extend from the frontal to the occipital and to the tem-
8
poral lobes, and are connected to the third ventricle by the foramen
of Monro. The aqueduct extends to the fourth ventricle and thus
connects supratentorial to infratentorial ventricles. The foramina of
Luschka and the foramen of Magendie are the (infratentorial) path-
ways between inner and outer CSF spaces (Figure 2.2). Disorders of
3 CSF circulation are called hydrocephalus, a condition that can be
acute or chronic. Sudden blockade of the aforementioned foramina
or the aqueduct—​the bottlenecks of CSF circulation—​lead to acute
4 9 failure of CSF circulation and symptoms of herniation (Box 2.1).
Aqueductal stenosis, blockade of the foramina of Monro (colloid
cysts), Luschka, and Magendie may occur acutely by a tumour mass
5 or blood clotting after intraventricular haemorrhage (within few
2
hours) or chronically by congenital membranes or slowly growing
6 masses. Malresorption of CSF may occur as a consequence of high
CSF protein content, which can occur post-​haemorrhage (degrad-
10 ation of blood cells), post-​infection (meningitis/​ventriculitis), or
as exudate (tumour, e.g., vestibular schwannoma), and can lead to
acute, subacute, or chronic hydrocephalus. Normal pressure hydro-
Figure 2.1 Schematic view of intracranial spaces and brain parenchyma.
(1) telecenphalon, (2) cerebellum, (3) diencephalon, (4) mesencephalon, (5) pons,
cephalus is a chronic CSF circulation disorder, leading to typical
(6) medulla, (7) falx, (8) lateral ventricles, (9) tentorium cerebelli, and (10) foramen clinical symptoms (Hakim-​Trias), i.e., cognitive and gait disorders
magnum. as well as urinary incontinence (5, 6).

Blood
brainstem towards the foramen magnum. The cranial nerves III to The cerebral blood volume makes up 5% of the ICV, mainly con-
XII originate from the brainstem. The brain parenchyma has to be sisting of venous vessels. Cerebral blood flow (CBF) and associ-
considered a rather static volume that is easily adaptive to chronic ated cerebral blood volume (CBV) are regulated by mechanisms
compression up to a certain extent (in cases of slowly growing tu- influencing cerebral resistance (i.e., constriction or dilation of ves-
mours, such as meningiomas), but very sensitive to acute com- sels, static autoregulation) over time (dynamic autoregulation). The
pression such as intracranial bleeding. Volume expansion of the pressure reactivity index (PRx) is described as a correlation coef-
parenchyma might be caused by tumour, abscess, intracranial ficient between (e.g., ten-​second average) ICP and mean arterial
haemorrhage (ICH), or oedema. The latter can be focal (e.g., in blood pressure (MAP) over a time window (e.g., five minutes). In
cases of tumour, bleeding, infection, stroke, venous stasis) or gen- severe brain injury patients, impaired autoregulation contributes to
eral (e.g., in SAH, global infarctions, traumatic brain injury, or as- unfavourable outcome. CPP is calculated as a difference between
sociated with systemic diseases). A natural atrophy of the brain can the MAP at head level and the ICP. The concept of the ‘optimal
be observed during ageing, which provides more buffer space for CPP’ should be followed, since values beyond the CPP level opti-
volume expansion in elderly people. mizing cerebral autoregulation are associated with fatal outcome or
increased disability. The optimal CPP is between 50 and 95 mmHg,
Cerebrospinal Fluid
but it is patient-​and time-​dependent, and thus needs continuous
The CSF space makes up 10% of the ICV, equalling a total of 120–​200 monitoring. In cases of acutely elevated ICP, decreasing the IBV
ml. CSF is a low-​protein, glucose-​containing liquid that contains is a short-​term strategy to decrease ICP. This can be achieved by
few cells, mostly lymphocytes (up to 4/​µl). It is mainly produced by moderate (short-​term) hyperventilation leading to hypocapnia
and associated cerebral vasoconstriction. Special caution has to be
undertaken in patients suffering from vasospasm after SAH.
Table 2.1 The contents of the cranial vault are divided into three
compartments. Proportions of intracranial volume are given in %.
Intracranial Compliance
Contents of the intracranial vault The pressure-​volume relationship between ICP, ICV, and CPP
pressure is known as the Monro-​Kellie doctrine, which states that
Brain parenchyma 85% i) the brain is enclosed in the non-​expandable cranium; ii) brain
Cerebrospinal fluid 10% parenchyma is nearly incompressible; iii) the blood volume of the
Blood 5% intracranial vault is nearly constant; and iv) a continuous outflow of
venous blood from the intracranial vault is required to make room
91

Chapter 2 intracranial pressure 19

increased ICP include continued decrease in level of conscious-


ness (stupor, coma), dilated pupils, no reaction of pupils to light,
vomiting, bradycardia, hyperthermia, and papilloedema. The ability
of the craniospinal space to accommodate for changes in ICV (CSV,
parenchyma, blood) is defined by a non-​linear hyperbolic relation-
ship between pressure and volume. Notably, ICP physiologically
can increase or decrease in return of thoracic pressure changes
(e.g., 2–​4 mmHg) during respiration. Head-​up positioning de-
1 creases ICP, as the pressure gradient between CSF and the venous
2 blood system increases. Special procedures may increase ICP (e.g.,
3 Trendelenburg positioning).

Cerebral Perfusion Pressure


4 Intracranial volume is regulated by the crystalloid osmotic pressure
(about 5000 mmHg) gradient across the impermeable blood-​brain
barrier (BBB). Whenever the BBB is severely damaged, this crystal-
loid osmotic gradient might be considerably small. Notably, colloid
pressure (about 20 mmHg) and hydrostatic pressure also account
for entry of water into brain parenchyma. It is important to main-
tain plasma crystalloid osmotic pressure and oncotic pressure in
case of acute brain injury. ICV, and thereby ICP, is predominantly
influenced by arterial partial pressure of carbon dioxide (PaCO2),
Figure 2.2 Structures of the CSF space.
(1) foramen of Monroi, (2) aqueduct, (3) prepontine cistern/​floor of the third
as CBF increases 2–​6% per mmHg increase in PaCO2 levels. Also,
ventricle, and (4) outlet of the 4th ventricle/​foramen Magendi. CBF is tightly coupled to cerebral metabolism and increases with
the increase in cerebral metabolic rate.
CPP is defined as CPP = MAP–​ICP. Normal CBF remains constant
for incoming arterial blood. Intracerebral compliance (CIC = ∆V/​ and ranges from 45–​50 ml/​100g brain tissue/​min. Cerebral perfusion
∆P) reflects the ability of the intracranial system to compensate and autoregulation may be disturbed in acute brain injury, poten-
for changes in volume (∆V) per unit change in pressure (∆P). tially causing an increase in CBV and ICP, thereby decreasing CPP
Intracerebral elastance (EIC = ∆P/​∆V) is the inverse of compliance and causing subsequent brain injury. Normal CPP values range be-
(Figure 2.3). tween 70 and 90 mmHg. In neurosurgical care, the lower therapeutic
ICP is measured at the level of the foramen of Monro. The thresholds are between 50 and 70 mmHg in traumatic brain injury
normal value for ICP at rest is 10±5 mmHg for a supine adult. Mild and 80 mmHg and more in special cerebrovascular pathologies.
ICP elevation is defined as 16–​20 mmHg, moderate ICP eleva-
tion as 21–​30 mmHg, and severe ICP elevation as 31–​40 mmHg
(1 mmHg = 0.133 kPa, see Table 2.2). Early clinical signs of in-
ICP Waveform
creased ICP include decreased level of consciousness, confu- The ICP pulse wave is a dynamic, pulsatile waveform that shows
sion, restlessness, lethargy, cerebral and pupillary dysfunction, oscillating components at two different frequencies (cardiac and
deterioration of motor function, headache, personality changes, respiratory cycles, see Figure 2.3). Normally, patients show a low
and decreasing Glasgow Coma Scale score. Late clinical signs of and stable ICP (<20 mmHg) waveform that fluctuates in response
to MAP variations. Additionally, ICP may fluctuate in response
to daily activities such as exercise or coughing (increase up to
110 mmHg). The normal ICP waveform consists of three charac-
Box 2.1 Clinical Alert Signs Indicating Brainstem and Eloquent teristic upstrokes (P1–​3). The first large peak P1 (percussion wave)
Cortical Area Herniation: Early Counteraction Required is generated by pulsations of major intracranial arteries and the
choroid plexus. The second smaller peak P2 (tidal wave) depends
◆​ Headache upon the intracranial elastance. The third peak P3 (dicrotic wave)
◆​ Neurological dysfunction is produced by aortic valve closure. ICP overall pulse wave can be
increased by expiration (increase in central venous pressure) and
◆​ Ipsilateral dilation of pupil
decreased by inspiration (decrease in central venous pressure).
◆​ Oculomotor paresis Changes in mean ICP (>20 mmHg), amplitude, and periodicity of
◆​ Hemiparesis pulsatile components might indicate reduced intracranial elastance.
For example, increase in P1–​3 amplitude might represent increased
◆​ Contralateral dilation of pupil
CSF volume. In contrast, when a large volume of CSF is drained off,
◆​ Pathological breathing or in the case of an incompletely closed skull after craniectomy, the
◆​ Bradycardia, hypertension ICP waveform will decrease in amplitude. A prominent P1 wave
may occur when the systolic blood pressure is very high. A dimin-
◆​ Apnoea ished P1 wave might occur when the systolic blood pressure is too
02

20 Section 1 neuroscience in anaesthetic practice

(a) 14 P2
13
12
11

ICP (mmHg)
10
9 P1
8
7
6
5
4
3
0 50 100 150 200 250 300 350 400
Time (ms)
(b) 14
P2
13
12
11 P1
P3

ICP (mmHg)
10
Pressure

9
8
7
6
5
4
3
0 50 100 150 200 250 300 350 400
∆p Time (ms)
(c) 10 P1
9

ICP (mmHg)
8 P2
7 P3
6
5
∆p
4
3
∆V Volume ∆V 0 50 100 150 200 250 300 350 400
Time (ms)

Figure 2.3 Cerebral curve (non-​linear hyperbolic relationship).


Initially, an increase in the ICV results to small increase in ICP. With increasing ICP, however, the same amount of increase in volume leads to a larger increase in ICP,
thus indicating reduced cerebral compliance (left). With increasing ICP there are typical changes of the ICP curve (right diagram; C, normal, P1>P2>P3; B, moderate
impairment, P2>P1>P3; A, severe impairment, P2 only, no P1/​P3).

low, leaving only P2, although P2 and P3 are not changed by this. changes, for example, during intracerebral vasodilation or
A prominent P2 wave may occur when intracerebral compliance vasoconstriction.
has decreased (e.g., increasing ICP) or during inspiratory breath
hold. During hyperventilation, P2 and P3 waves might diminish. Lundberg A–​C Waves
A rounded ICP waveform with reduced peaks from P1–​3 might
occur when ICP is critically high. A number of pathological waves Lundberg A-​waves (plateau waves) are clearly pathological and de-
have been described so far. Modern extended ICP pulse waveform fined as a sudden ICP increase up to 50–​100 mmHg lasting 5–​20
analysis (e.g., morphological clustering and analysis of ICP, or minutes (Figure 2.4). During A-​waves, it is common to develop
MOCAIP) potentially may allow for detection of cerebrovascular clinical signs and symptoms of early herniation, including brady-
cardia and hypertension. Although the underlying mechanism
remains unknown, it has been postulated that CPP cannot meet
cerebral metabolic demand, thereby triggering cerebral vasodila-
Table 2.2 Typical ICP values during different activities and sleep tion and subsequent increase in CBV and ICP. This, in return,
in babies and adults. causes additional CPP decrease, ultimately resulting in a vicious
cycle. Atypical Lundberg A-​waves do not exceed an elevation of
Activity Baby [mmHg] Adult [mmHg] 50 mmHg and are an early indicator of neurological deterioration.
Lying supine 6±1 10±5 Lundberg B-​waves are oscillating waves defined as a short modest
ICP increase of 10–​20 mmHg lasting 0.5–​2 minutes. It has been
Standing up –​5±5 –​5±5
postulated that B-​waves might be caused by vasomotor centre
Non-​REM-​sleep 7±2 12±5 instability when CPP is unstable or at the lower limit of cerebro-
REM-​sleep 19–​22 15–​25 vascular autoregulation. Lundberg C-​waves are rapid sinusoidal
fluctuations of up to 20 mmHg of ICP lasting for 7–​15 seconds
Coughing, sneezing 20–​40 30–​110
(about 0.1 Hz). These waves correspond to Mayer fluctuations in
12

Chapter 2 intracranial pressure 21

Lundberg A waves
60
50
40

ICP (mmHg)
30
20
10
0
0 10 20 30 40 50 60 70 80
Time (min)
60 Lundberg B waves
50
ICP (mmHg)

40
30
20
10
0
0 10 20 30 40 50 60 70 80
Time (min)
60
Lundberg C waves
50
ICP (mmHg)

40
30
20
10
0
0 10 20 30 40 50 60 70 80
Time (min)

Figure 2.4 Lundberg A waves (5–​50 mmHg, 5–​20 minutes), Lundberg B waves (10–​20 mmHg, 0.5–​2 minutes) and Lundberg C waves (several mmHg, about 0.1 Hz).

MAP that have been documented in healthy individuals and poten- in terms of mortality. When measured over time at different CPP
tially are caused by cardiovascular interactions. thresholds, PRx demonstrates a U-​shaped curve, suggesting a spe-
cific relationship of individual autoregulation. This approach can
be used to tailor individual CPP management in order to optimize
Cerebrovascular Pressure Reactivity the patient’s autoregulation. As ABP and ICP routinely are meas-
CBF autoregulation describes the ability of the cerebral vasculature ured continuously in patients at risk, this index is readily available
to maintain a stable CBF despite fluctuations in CPP. With intact when using appropriate software. Notably, the underlying prin-
autoregulation, a rise in arterial blood pressure (ABP) produces ciple of PRx also works for cerebral variables other than the ICP,
cerebral vasoconstriction, a decrease in CBV, and a fall in ICP (7, such as cross-​correlating MAP and CBF velocity determined by
8). A fall in ABP produces the opposite effects. With disturbed transcranial Doppler sonography (Mx) or regional frontal haemo-
autoregulation, a rise in ABP is transmitted to the intracranial com- globin oxygen saturation determined by near-​infrared spectros-
partment and produces a rise in ICP as a passive pressure effect. copy (ORx, THx) ideally determined at high temporal resolution.
Cerebral autoregulation can be determined by investigating the
moving Pearson correlation coefficient between changes in ABP ICP Measurement
and ICP, i.e., PRx. In fact, waveforms of ABP and ICP are correl-
ated at higher temporal resolution. PRx is negative (e.g., between Intraventricular Catheters
0 and –​1) when cerebral vessels are pressure reactive and aim to Direct measurement of ICP in the lateral ventricle is still con-
counteract changes in ABP. Instead, PRx is positive (e.g., between sidered the ‘gold standard’ for ICP measurement (Figure 2.5 and
0.3 and 1) when cerebral vessels are not pressure reactive and alter- Box 2.2) (9). The measurement is performed at the level of the me-
ations in MAP are mostly directly transmitted to ICP. PRx has been atus acusticus externus corresponding to the foramen of Monro.
identified as a predictor of outcome after traumatic brain injury Advantages of this measurement include the possibility of external
2

22 Section 1 neuroscience in anaesthetic practice

Figure 2.5 Intraventricular and intraparenchymal ICP measurement.

calibration and CSF drainage. However, placement of the catheter ventricular drain (EVD) or a lumbar drain (LD). The latter can only
may be difficult or even impossible in case of severe brain swelling be applied in communicating hydrocephalus, and an open passage
with collapsed ventricles. Furthermore, the risk of infection is re- through the aqueduct and the outlet of the fourth ventricle is man-
ported to be between 3.5% to more than 20% according to different datory, which can be estimated by radiologic imaging (i.e., to check
larger clinical studies. Many authors report an increase of infection for mass lesions and ventricular diameter gradients). Blockage of
risk with prolonged usage (10). In clinical practice it is important to CSF pathways could lead to transtentorial or transforaminal her-
be aware that ICP recordings are representative only with a closed niation (see Herniation Syndromes).
drainage system (4, 11, 12, 13, 14). In many cases, an external ventricular drainage is the first choice.
It can be implanted in a sterile surrounding at the intensive care
Intraparenchymal Probes unit (ICU) or in the operating theatre by a neurosurgeon and com-
ICP can be recorded by inserting a probe into the brain paren- bines draining of CSF with measurement of ICP. The EVD is a sili-
chyma. Usually, these probes are inserted in a non-​eloquent brain cone catheter that is inserted into the lateral ventricle, usually by a
area, although the exact placement, especially in focal pathologies, frontal and rarely by an occipital or temporal approach, through a
is a matter of ongoing debate. Measurement of ICP is local and does burr hole and a small incision of the dura (15, see Table 2.3). The
not necessarily represent CSF pressure. The risk for parenchymal most common variant is a frontal burr hole over Kocher’s point,
bleeding or infection is <1%. Microtransducers cannot be recali- which is 1 cm anterior to the coronary suture and 3 cm lateral from
brated after insertion and drift from zero might occur during long-​ the midline (Figure 2.6). The target point is the entrance of the for-
term measurement. amen of Monro which is situated at the crossing of virtual lines
through the medial canthus of the eye (anterior-​posterior) and the
Drainage of CSF external acoustic meatus (left-​right). The EVD can be attached to
the bone via a bolt system or tunnelled subcutaneously. The ven-
In cases of acute hydrocephalus, drainage of CSF has to be per- tricular catheter is secured by suturing it to the skin, although it
formed promptly. This can be done by insertion of an external may also be connected to a second distal silicone catheter with a
‘Vienna connector’ to prevent accidental removal by the patient or
during nursing procedures. The CSF drainage system includes a
Box 2.2 Reasons for Performing ICP Monitoring drip chamber that is positioned according to the patient’s head, at a
certain level above reference points such as the radix nasi or the ex-
◆​ Severe brain trauma (GCS 3–​8) ternal acoustic meatus (point where the tip of the ventricular cath-
eter is estimated to be, i.e. the opening of the foramen of Monro).
◆​ Severe subarachnoid haemorrhage (traumatic, vascular) The level of the drip chamber can be adjusted according to a re-
◆​ Severe cerebral ischaemia sistance for the drained CSF target amount (principle of commu-
nicating vessels). It is typically placed between 0 and 15 cm above
◆​ Diagnostic (hydrocephalus, shunt insufficiency)
the reference points and adapted to ICP (normal values between
32

Chapter 2 intracranial pressure 23

Table 2.3 Step-​by-​step guide for the implementation of an external


ventricular drain inserted into the lateral ventricle via a burr hole
over Kocher’s point (see Figure 2.5 for anatomical landmarks).

1 Define Kocher’s point (3 cm lateral from the midline, 1 cm


anterior from the coronary suture)
2 Define skin incision for drainage outlet
3 Shaving of the head
4 Washing with disinfectant, marking of Kocher’s point, drainage
outlet, and a 3 cm incision (in the course of a possible skin
incision if microsurgical operation is planned on this side) + sterile
draping
5 Skin incision, lifting of periosteum, insertion of a spreader,
coagulation with bipolar cautery
6 Burr hole
7 Coagulation of dura
8 Incision of dura (rather small to prevent CSF fistula)
9 Coagulation of superficial cortex
10 Placement of ventricular catheter (orthogonal to the bone,
target as described); depth measured on CCT scan before hands
(mostly 5.5–​6 cm from dura; should be marked on catheter);
the ependymal lining is felt as a tactile resistance; the catheter is
only inside the ventricles when CSF is flowing (catheter end can
be placed as low as possible to enhance flow); CSF probes to
bacteriology and cell count
11 Temporary clamping of catheter (bulldog clamp) as close to the Figure 2.6 Axial view of the lateral ventricles in reference to the skull.
dura as possible—​marks depth The vertical bar crosses the medial angle of the eye, the horizontal bar represents
a virtual connection of both external acoustic meatus—​the crossing marks the
12 Incision at skin outlet target structure for the tip of the EVD, i.e., the Foramen of Monro. The arrow tip
points towards Kocher’s point marking the entry zone of the EVD. It is situated
13 Tunnelling of catheter (still clamped)
1 cm anterior to the coronal suture, 3 cm paramedian.
14 Testing of catheter function
15 Skin closure with subcutaneous and skin sutures (bulldog clamp
removed and placed at catheter end) plastic tube. The distal drainage system may be blocked by blood
16 Fixation of catheter on skin with connector remnants and can be changed under sterile conditions. The prox-
imal catheter may be tilted or clotted. Furthermore, attached valves
17 Connection of ‘Vienna connector’ system must be checked. Aspiration of CSF can be performed in order to
18 Tube connector mobilize blood clots from inside the silicone tube. Sterile condi-
19 Testing of catheter function tions have to be guaranteed, and the ‘no-​touch technique’ should
be used during the procedure. All connectors should be cleaned
20 Connection to valve and distal collection system with disinfectant before attaching a syringe. Only syringes up to
21 Skin closure 2 ml of volume should be used in order to avoid excessive negative
pressure at the tip of the silicone catheter. Plexus choroideus, epen-
NB: ICP probes and EVD probes with bolt systems should be implanted as described by
the respective manufacturers. dyma, and brain tissue could be aspirated, leading to injury of brain
tissue or intraventricular haemorrhage. Aspiration of CSF should
only be performed by experienced team members. Knowledge of
0 and 20 mmHg), flow volume (10–​25 ml/​hour), and ventricular the width of the lateral ventricles is mandatory for this procedure. If
size. Lowering of the drip chamber below the level of the foramen of the ventricles are totally collapsed (slit ventricles) by CSF drainage
Monro (including dropping it) may lead to serious over-​drainage of or cerebral oedema, aspiration is dangerous and should not be
CSF and, if unnoticed, may result in upwards transtentorial hernia- performed.
tion. The position of the ventricular catheter may be controlled by a Flushing of the silicone catheter is an even more invasive pro-
computed tomography (CT) scan. The EVD may stop draining CSF cedure and should be performed only by experienced team mem-
due to several reasons, all of which must be checked step by step in bers after careful consideration. The risk of introducing germs into
order to prevent malfunctioning and increase of ICP. The ICP may the ventricular system is very high when not performed under
be below the counter-​pressure maintained by the level of the drip sterile conditions. All connectors have to be disinfected multiple
chamber. Functioning of the EVD can easily be checked by lowering times before attachment of a syringe filled with 0.9% sodium
the drip chamber for a few seconds, when the CSF should again chloride solution or artificial CSF. No more than 1–​2 ml should be
start to drain and CSF pulsation can be seen inside the transparent injected into the silicone catheter. If all these procedures fail to start
42

24 Section 1 neuroscience in anaesthetic practice

the EVD functioning, a cranial computed tomography (CCT) scan compression of the oculomotor nerve. This complements to the
should be considered to check for ventricular size or dislocation stretching of the nerve over the clivus (due to downward shift of
of the ventricle catheter. The ICP should only be measured by the its origination), resulting in ipsilateral mydriasis at an early stage.
transducer when the distal drainage system is closed. A lumbar CSF This is an absolute alert sign—​immediate lowering of ICP has to
drain is inserted intradurally under sterile conditions between ver- be achieved.
tebrae L4/​5, L3/​4, or L2/​3. The level of L4/​5 can easily be found by ◆ Subfalcine: Parts of the hemisphere inheriting the mass lesion
connecting the palpable rims of the anterior iliac crests. The drain may herniate below the falx cerebri, possibly compressing the
is also connected to a drip chamber and a distal drainage system. pericallosal and callosomarginal arteries, leading to brain in-
However, ICP measurement is not reliable. From both types of farcts of the medial frontal lobe (gyrus cinguli, gyrus frontalis
drainages, CSF should be analysed at regular intervals (e.g., every superior) down to the precuneus. On CCT, a midline shift can
second day). The following parameters may be evaluated (Table be seen (measured at the level of the septum pellucidum or the
2.4). Additionally, microbiological cultures should be performed pineal gland). Greater than 5 mm of midline shift may lead to
regularly. stupor, and greater than 9 mm to coma.
Transtentorial: When the supratentorial ICP increases, structures
Herniation Syndromes ◆
shift further downwards, leading to transtentorial herniation. It is
There are different reasons for causes of impairment of conscious- a rather late, often terminal event, leading to direct compression of
ness in neurological and neurosurgical patients, and conscious- the brainstem. The oculomotor nuclei are damaged on the contra-
ness is dependent on the functioning of the ascending reticular lateral side, resulting in mydriasis and loss of light reaction of the
activating system (ARAS), a network of neurons with connections contralateral (in addition to the ipsilateral) pupil. Bilateral mydriasis
from the pons up to the cerebral cortex. The following three terms and decerebrate posturing (flexion of upper, extension of lower ex-
are often used to describe impairment of consciousness, but all tremities) are signs of a late phase of herniation. Furthermore, the
terms used should be complemented by detailed clinical descrip- posterior cerebral artery (PCA) is at risk—​its compression leads to
tions of the patient. parietooccipital brain infarcts (visual impairments, blindness).
1) Somnolence is a sleep-​like state that can be disrupted easily by ◆ Tonsillar: Raised pressure in the posterior fossa leads to her-
speaking to the patient. Wakefulness and awareness are im- niation of the cerebellar tonsils through the foramen magnum.
paired and attention cannot be sustained. Compression of the medulla oblongata leads to respiratory arrest
2) Stuporous patients may be roused only by strong stimuli, such and compression of the outflow of the fourth ventricle to acute
as pain. hydrocephalus.

3) Coma is characterized by a loss of consciousness with an Besides impairment of consciousness, other signs of increasing ICP
unarousable patient, even to strong stimuli. Motor responses have to be considered, such as headache, altered brain function
are possible. (sensory or motor deficits, speech impairment, cognitive dysfunc-
tion), seizures, and visual disturbances. Conservative measures
Different grading scales such as the Glasgow Coma Scale or the Full should be undertaken before aiming at surgical interventions (Box
Outline of Unresponsiveness score are used to describe these states 2.3). There should at least be a consideration of surgical intervention
in more detail. Increase of ICP leads to compression of the struc- for intracranial lesions with mass effect leading to clinical symp-
tures relevant for the ARAS by ‘herniation’ through the tentorial toms. Extra-​axial and intra-​axial tumours, intracranial bleedings
notch and the foramen magnum or the facial and tentorial edges: (especially epidural and subdural haematoma, but in some cases
◆ Uncal and Central: Unilateral, supratentorial mass lesions shift also intracerebral haemorrhage), as well as large abscesses often re-
the brain downwards and towards the contralateral side, leading quire surgical intervention. Mass lesions in the posterior fossa—​
to compression of the diencephalon and the brainstem in the including intraparenchymal bleeds—​have to be surgically treated
tentorial notch and at the tentorial edge, thus leading to im- when compression of the fourth ventricle is suspected or expected
pairment of consciousness. Also, temporal structures, especially due to swelling. In cases of strokes, conservative management
the uncus, herniate around the tentorial edge, leading to direct should be aimed at, except from large cerebellar strokes. However,

Table 2.4 The following parameters of the cerebrospinal fluid are important for the differential diagnosis of infection and haemorrhage.

CSF Compound Normal Values Pathological Changes


Cell count 1–​4/​µl up to 100s + activated lymphocytes = reactive (viral) meningitis; 1000s + neutrophil
granulocytes = bacterial meningitis.
Glucose >50% of serum glucose <50% of serum glucose = bacterial growth.
Protein <100 mg/​dl High protein in tumours, bacterial meningitis, SAH.
Lactate <1–​2 mmol/​L High lactate in SAH or bacterial growth.
Cytology Few lymphocytes, monocytes Activated lymphocytes in viral meningitis; Neutrophil granulocytes in bacterial
meningitis; siderophages (12h to >3d), erythrophages (<12h to 3d) in SAH/​IVH.
52

Chapter 2 intracranial pressure 25

4. Le Roux P, Menon DK, Citerio G, Vespa P, Bader MK, Brophy G,


Box 2.3 Management of Increased ICP et al. The International Multidisciplinary Consensus Conference
on Multimodality Monitoring in Neurocritical Care: Evidentiary
In cases of increased ICP, it is vital to optimize parameters
tables: A statement for healthcare professionals from the Neurocritical
of ventilation, haemodynamics, sedation, and positioning. Care Society and the European Society of Intensive Care Medicine.
Additionally, both conservative and surgical therapy options Neurocritical Care. 2014;Suppl 2:S297–​361.
should be considered. 5. Fishman RA. Cerebrospinal fluids in diseases of the nervous system.
Philadelphia, PA: Saunders; 1992.
◆​ Optimization of PaCO2 (35 mmHg), PaO2 (120 mmHg), tem-
6. Rosenberg GA. Brain fluids and metabolism. Oxford: Oxford University
perature (<37.5°C/​99.5°F), serum electrolytes (Sodium), pH. Press; 1990.
◆​ Drainage of CSF, check for 30° head up and optimal cranial 7. Kety SS, Shenkin HA, Schmidt CF. The effects of increased intracranial
venous outflow. pressure on cerebral circulatory functions in man. The Journal of
Clinical Investigation. 1948;27:493–​9.
◆​ Identify potential causes for increased ICP, consider CCT. 8. Kirkman MA, Smith M. Intracranial pressure monitoring, cerebral
◆​ Consider volume and vasopressor in case of hypotension for perfusion pressure estimation, and ICP/​CPP-​guided therapy: A
standard of care or optional extra after brain injury? British Journal of
individual target CPP. Anaesthesia. 2014;112:35–​46.
◆​ Consider osmodiuretics (e.g., mannitol 20% 125–​250 ml, 9. Narayan R, Kishore P, Becker DP, Ward JD, Enas GG, Greenber RP,
cave: plasma osmolality). et al. Intracranial pressure: To monitor or not to monitor? A review of
our experience with severe head injury. The Journal of Neurosurgery.
◆​ Consider temporary (5–​10 min) hyperventilation (e.g., PaCO2 1982;56:650–​9.
30 mmHg). 10. Pfisterer W, Mühlbauer M, Czech T, Reinprecht A. Early diagnosis
of external ventricular drainage infection: Results of a prospective
◆​ Consider neurosurgical therapy (e.g., decompressive
study. The Journal of Neurology, Neurosurgery & Psychiatry.
hemicraniectomy). 2003;74:929–​32.
◆​ Consider barbiturate coma (e.g., thiopental bolus 5–​10 mg/​ 11. Chesnut R, Videtta W, Vespa P, Le Roux P; Participants in
kg or 3–​5 mg/​kg/​h) or mild hypothermia (e.g., 33–​35°C/​ the International Multidisciplinary Consensus Conference
on Multimodality Monitoring. Intracranial pressure
91.4–​95°F).
monitoring: Fundamental considerations and rationale for monitoring.
Neurocritical Care. 2014;Suppl 2:S64–​84.
12. Citerio G, Oddo M, Taccone FS. Recommendations for the use of
when brain shift increases, decompressive hemicraniectomy within multimodal monitoring in the neurointensive care unit. Current
48 hours after onset leads to favourable outcomes in patients Opinion in Critical Care. 2015;21:113–​9.
13. Czosnyka M, Miller C; Participants in the International
younger than 60 years (adapted from 16).
Multidisciplinary Consensus Conference on Multimodality
Monitoring. Neurocritical Care. 2014;Suppl 2:S95–​102.
References 14. Helbok R, Olson DM, Le Roux PD, Vespa Paul; Participants in
the International Multidisciplinary Consensus Conference on
1. Cushing H. The blood-​pressure reaction of acute cerebral compression,
Multimodality Monitoring. Intracranial pressure and cerebral
illustrated by cases of intracranial hemorrhage. American Journal of
perfusion pressure monitoring in non-​TBI Patients: Special
Medical Sciences. 1903;125:1017–​45.
considerations. Neurocritical Care. 2014;Suppl 2:S85–​94.
2. Lundberg NG. Continuous recording and control of ventricular
15. Marcus HJ, Wilson MH. Videos in clinical medicine. Insertion of
fluid pressure in neurosurgical practice. [Doctoral dissertation]
an intracranial-​pressure monitor. New England Journal of Medicine.
Copenhagen: Ejnar Munksgaard; 1960.
2015;373:e25.
3. Lundberg N, Troupp H, Lorin H. Continuous recordings of the ventricular-​
16. Koenig M. Cerebral herniation syndromes and intracranial hypertension.
fluid pressure in patients with severe acute traumatic brain injury.
Updates in neurocritical care. New Brunswick, NJ: Rutgers University
A preliminary report. The Journal of Neurosurgery. 1965;22:581–​90.
Press; 2016.
62
Another random document with
no related content on Scribd:
120 Tab. X, 4–6 (Schädel); id. 1893 III, 78; W e b e r 1894 ib.
474.
Mus chrysocomus („n. sp. 3o“ J e n t i n k 1879 T. Ned. D. Ver. IV
p. LVI); B. H o f f m a n n 1887 Abh. Ber. Dresd. 1886/7 Nr. 3 p.
20, Tafel Fig. 1 a–f (Schädel); T h o m a s 1895 AMNH. (6) XVI,
163 und 1896 XVIII, 247; T r o u e s s a r t 1897 Cat. Mam. 485;
T h o m a s 1898 TZS. XIV, 403.
Mus fratrorum T h o m a s 1896 AMNH. (6) XVIII, 246;
Trouessart 1897 Cat. Mam. 485.
fem.,
a, b.
in Spiritus, Tomohon, Minahassa, Nord Celébes, III
und IV 94.
mas juv.,
c. in Spiritus, Tomohon, III 94.
fem.
d, e.
juv., in Spiritus, Tomohon, III 94.

Als H o f f m a n n 1887 M. chrysocomus beschrieb, besass das


Dresdner Museum kein Exemplar von callitrichus, er war auf
J e n t i n k s Beschreibung angewiesen. 1894 trafen aber 4
Exemplare ein, die Dr. J e n t i n k die Güte hatte, mit seinen Typen
zu vergleichen und als solche zu bestimmen, man kann daher an
ihrer Identität nicht zweifeln, trotzdem die Beschreibung der Art
(NLM. 1879 I, 12) nicht so zutreffend und genügend ist, dass sie
danach allein sicher erkannt werden könnte. Zwischen dem einzig
vorhandenen Typus von chrysocomus und den mir nun vorliegenden
Exemplaren von callitrichus kann ich aber keine irgendwie
wesentlichen Unterschiede constatiren, sowenig wie zwischen M.
fratrorum Thos. (wovon das Dresdner Museum 2 von T h o m a s
bestimmte Exemplare besitzt) und callitrichus. Dieser sagt (AMNH.
XVIII, 247), dass fratrorum M. chrysocomus sehr nahe stehe, aber
durch Grösse, geperlte Supraorbitalränder und mächtigere Molaren
unterschieden sei, allein die Schädel der zwei mir vorliegenden
Exemplare zeigen diese Perlung nicht, sondern haben scharfe
Ränder wie gewöhnlich; Grösse und mächtigere Molaren können als
Artunterschiede, in Ansehung der bedeutenden Differenzen der
Exemplare nach Alter und Geschlecht, nicht angesehen werden.

J e n t i n k identificirte ferner einen Schädel ohne Unterkiefer von


Parepare, Süd Celébes (Webers Zool. Erg. I, 120) mit callitrichus
und sagt, dass es sehr leicht sei, die Art nur nach dem Schädel zu
unterscheiden, unterlässt es aber die unterscheidenden Charaktere
anzugeben; er verweist nur auf einige Abbildungen zum Vergleiche
(Cat. MPB. IX Pl. 7, Zool. Erg. I Tab. X), die aber hierfür, in
Ansehung der beträchtlichen Unterschiede nach Alter und
Geschlecht und wegen der nicht hinlänglich deutlichen Details an
den Zähnen, nicht genügen. Ich halte eine solche Identificirung für
unsicher und möchte erst weiteres Material von Süd Celébes
abwarten, so wenig ich die Möglichkeit des Vorkommens von M.
callitrichus in Süd Celébes in Abrede stellen will. [25]

Endlich hat T h o m a s neuerdings (TZS. XIV, 403) M. chrysocomus


vom Berge Data, Lepanto, Nord Luzon, von 8000 Fuss Höhe
aufgeführt und bemerkt, dass die Art von fast allen anderen der
Gattung durch das völlige Fehlen der scharfen Supraorbitalränder
unterschieden sei. Ein von T h o m a s bestimmtes, ebenfalls
männliches, ziemlich adultes Exemplar im Dresdner Museum von
demselben Fundorte zeigt am Schädel ebensowenig scharfe
Supraorbitalränder, während der Typus von chrysocomus von Nord
Celébes, ein noch junges Exemplar, diese deutlich markirt hat, wie
auch aus der H o f f m a n n schen Abbildung ersichtlich ist, und wie
es der von mir angenommenen Identität mit callitrichus entspricht.
Da nun ausserdem das Exemplar von Luzon einen viel weicheren
und nicht so lebhaft gefärbten Pelz hat wie callitrichus (und
chrysocomus) und noch andere kleine Unterschiede aufweist, so
möchte ich, auch unter Berücksichtigung des entlegenen und hohen
Fundortes, trotz notorisch vorhandener Ähnlichkeiten, die Identität
nicht vertreten und nenne die Exemplare vom Berge Data: Mus
datae. Erst bei einer weit besseren Kenntniss der Mäuse dieser
Gegenden, die wohl noch lange auf sich warten lassen wird, kann
man zu einer klareren Einsicht, als es jetzt möglich ist, gelangen.

Was die speciellen Fundorte von M. callitrichus auf Celébes angeht,


so ist die Art im Norden aus der Minahassa registrirt von Manado,
Langowan, Kakas (Mus. Leid.), Tomohon (Sarasins), Lotta (Mus.
Dresd.), Rurukan 3500 Fuss hoch („fratrorum“ Brit. Mus. und Mus.
Dresd.), Amurang („chrysocomus“ Mus. Dresd.); im Süden von
Parepare, welcher letztere Fundort aber meiner Ansicht nach noch
der Bestätigung bedarf.

[Inhalt]

36. Mus hellwaldi Jent.

Tafel VII Fig. 2–10

J e1879
n t i n k T. Ned. D. Ver. IV p. LV („5o“); id. NLM. I, 11
id.1883
ibid. V, 176
id.1887
Cat. MPB. IX, 212
id.1888
ibid. XII, 65
W1894
e b e r Zool. Erg. III, 474
T 1896
h o m a s AMNH. (6) XVIII, 246
T r1897
o u e s s a r t Cat. Mam. 479.
mas,a. in Spiritus, Minahassa, Nord Celébes, 8. V.

Bis jetzt nur von der Minahassa bekannt: Manado, Langowan,


Amurang (Mus. Leid.), denn die Angabe, dass die Art auch auf
Bórneo und Bunguran (Natuna Ins.) vorkäme, hat T h o m a s
(AMNH. 6. s. XIV, 455 1894) zurückgezogen (vgl. auch H o s e Mam.
Borneo 1893, 59, Nov. Zool. I, 658 1894 und NLM. XIX, 160 1897).

Der Färbung und weichen Beschaffenheit des Pelzes nach eine sehr
schöne Art. Der Schwanz ist (nach dem S a r a s i n schen Spiritus-
Exemplar) unten gelblich, oben im basalen Drittel grau, im mittleren
zu gelblich übergehend, im distalen gelblich wie unten (dies zur
Ergänzung der J e n t i n k schen Beschreibung). Die schöne braune
Farbe der Oberseite ist scharf von der weissen Unterseite abgesetzt,
auch an den Beinen.

Bezüglich der einzelnen Figuren siehe die Tafelerklärung.

[Inhalt]

37. Mus xanthurus Gr.

Tafel VI Fig. 2–10

J.1867
E. G r a y PZS. 598
G1879
ü n t h e r ib. 75 (Mus everetti); J e n t i n k T. Ned. D.
Ver. IV p. LV („4o“) und p. LVI („2o“); id. NLM. I, 10
J e1883
n t i n k NLM. V, 177
H 1887
o f f m a n n Abh. Ber. Dresd. 1886/7 Nr. 3 p. 1, 4, 13;
T h o m a s PZS. 514 (Mus xanthurus und everetti);
J e n t i n k Cat. MPB. IX, 212
J e1888
n t i n k Cat. MPB. XII, 66
H 1893
i c k s o n Nat. N. Cel. 229 [26]
W1894
e b e r Zool. Erg. III, 474
T 1895
h o m a s AMNH. (6) XVI, 163 (Mus everetti)
id.1896
ibid. XVIII, 246
T r1897
o u e s s a r t Cat. Mam. 472
T 1898
h o m a s TZS. XIV, 400 (Mus everetti).
Bälge
a, b.mit Schädel, mares, Tomohon, Minahassa, Nord
Celébes, 11. VII 94 und 30. III 95.
Balg c.mit Schädel, fem. juv., Makassar, Süd Celébes. 26.
XI 95.
2 mares,
d–f. 1 fem., in Spiritus, Tomohon, Minahassa, Nord
Celébes, III und IV 94.
1 mas,
g, h. 1 fem., in Spiritus, Minahassa.
fem. juv.,
i. in Spiritus, Matinang Südspitze, 29. VIII 94.

An manchen Exemplaren ist das Schwanzende behaarter als bei


anderen, ein behaarteres ist Tafel VI, 2 abgebildet.

G ü n t h e r beschrieb M. everetti von Mindanao oder einer kleinen


Insel der Nachbarschaft; er hat zwar keinen Fundort angegeben,
während solche bei allen anderen Arten, die in der Abhandlung
vorkommen, nicht fehlen, allein da sie sich nur über Mindanao-
Thiere oder Thiere der nächsten Nachbarschaft verbreitet, so scheint
der Fundort nicht zweifelhaft; ebensowenig sagt T h o m a s (TZS.
XIV, 400), woher das Exemplar stammte, er erwähnt aber die Art
vom Berge Data, Nord Luzon, 7500 Fuss hoch, von wo auch das
Dresdner Museum ein Exemplar von derselben Ausbeute und
demselben Fundort erhielt. An diesem allein kann ich die Identität
feststellen, denn G ü n t h e r s Beschreibung ist ungenügend. Da
nun aber keine wesentlichen Unterschiede zwischen diesem
Exemplar und denen von M. xanthurus von Celébes vorhanden zu
sein scheinen und Mindanao (oder Nachbarschaft) die Brücke
zwischen Celébes und Luzon bildet, so ziehe ich sie zusammen bis
eventuell eine bessere Kenntniss mir Unrecht giebt.

Die Fundorte auf Celébes sind bis jetzt in der Minahassa: Tondano
(Brit. Mus.), Manado, Langowan, Tondano, Kakas, Amurang (Mus.
Leid.), Manado, Amurang, Lotta, Rurukan 3500 Fuss hoch, Berg
Masarang 3500 Fuss hoch (Mus. Dresd.), Berg Kelelonde 4000 Fuss
hoch (Hickson), Tomohon (Sarasins); ausserhalb der Minahassa:
Matinangkette, westlich vom Gorontaloschen (Sarasins); im Süden:
Makassar (Mus. Dresd. und Sarasins).

Die Art muss sehr zahlreich vorkommen nach der relativ grossen
Zahl von Exemplaren im Leidener und Dresdner Museum und in der
Ausbeute der Herren Sarasin zu urtheilen (in Dresden 17).
H i c k s o n erwähnt dies auch für den Berg Kelelonde und sagt,
dass diese Ratten die saftigen Stiele der Kaffeebeeren besonders
lieben und daher den Plantagen sehr schaden. Ratten sind in der
Minahassa eine gesuchte Zuspeise zum Reise, 3 Arten Rattenfallen
von dort befinden sich im Museum der Bataviaasch Genootschap
(Not. XXV, 145 1897 und LIV [1898]), was beides für die Häufigkeit
der Thiere spricht.

Bezüglich der einzelnen Figuren siehe die Tafelerklärung.

[Inhalt]

38. Lenomys meyeri (Jent.)

Tafel VIII. Nat. Grösse

J e1879
n t i n k T. Ned. D. Ver. IV p. LV („7o“) und LVI („5o“, J.
verwies hier irrthümlich auf 6o p. LV = M. callitrichus); id.
NLM. I, 12
H 1887
o f f m a n n Abb. Ber. Dresd. 1886/7 Nr. 3 p. 12, 17, 21,
Tafel Fig. 2 a und b (Zähne); T h o m a s PZS. 514;
J e n t i n k Cat. MPB. IX, 211 pl. VII, 5–8 (Schädel)
J e1888
n t i n k Cat. MPB. XII, 65
W1894
e b e r Zool. Erg. III, 474
T 1896
h o m a s AMNH. (6) XVIII, 246
T r1897
o u e s s a r t Cat. Mam. 472
T 1898
h o m a s TZS. XIV, 409 Anm., pl. XXXVI, 1 (Zähne).
Lenomys, von den früheren Autoren zu Mus gestellt.
Bälge
a, b.mit Schädel, fem., Tomohon, Minahassa, Nord
Celébes, 6. und 18. III 94.
Skelette,
c, d. mas, fem., Tomohon III 94. [27]
Skelet, e. mas, Kema, Minahassa, Nord Celébes, VIII 93.
mares,
f, g. in Spiritus, Tomohon, III 94.

Die Individuen variiren in der Färbung zwischen mehr Grau und


mehr Braun, auf Tafel VIII ist ein graueres Exemplar abgebildet.

J e n t i n k (NLM. I, 13) sagt, dass die braunen Schnurrhaare weiss


gespitzt seien, allein dies ist bei den mir vorliegenden 8 Exemplaren
(ausser den obigen noch 4 des Dresdner Museums) nicht der Fall,
höchstens dass man bei dem einen oder andern vielleicht eine
schwache Andeutung davon sehen könnte; keinenfalls ist diese
Angabe für die Art charakteristisch.

Bis jetzt nur aus der Minahassa und dem Gorontaloschen bekannt,
aus letzterem von Bone (Mus. Leid.), aus ersterer von Manado-
Langowan (Mus. Leid.), Lotta, Rurukan 3600 Fuss hoch, Berg
Masarang 3500 Fuss hoch, Amurang (Mus. Dresd.) und Tomohon
(Sarasins). Vielleicht ist der Verbreitungsbezirk der Art über Celébes
ein viel grösserer. Wenn man bedenkt, wie lange dieses relativ
grosse Thier aus der Minahassa, wo so viel gesammelt worden ist,
unbekannt blieb, so dürfte diese Vermuthung nicht ungerechtfertigt
erscheinen.
[Inhalt]

39. Craurothrix leucura (Gr.)

Tafel IX. Nat. Grösse

J.1867
E. G r a y PZS. 599 Echiothrix (Schädel abgebildet)
J e1879
n t i n k T. Ned. D. Ver. IV p. LVI. Echiothrix
id.1880
NLM. II, 12. Echiothrix
id.1883
ibid. V, 177. Echiothrix
id.1888
Cat. MPB. XII, 73. Echiothrix
F 1891
l o w e r & L y d e k k e r Intr. Mam. 477 Echinothrix
W1894
e b e r Zool. Erg. III, 474 Echiothrix
T 1896
h o m a s AMNH. (6) XVIII, 246 Craurothrix
T r1897
o u e s s a r t Cat. Mam. 502 Echiothrix.
Balg a. mit Schädel, fem., Tomohon, Minahassa, Nord
Celébes, 11. VII 94
2 fem.,
b–d. 1 mas juv., in Spiritus, Tomohon, IV 94 und IV 95.

Bis jetzt nur von der Minahassa, Nord Celébes, bekannt, und zwar
von den Localitäten Amurang (Mus. Dresd. und Mus. Leid.), Berg
Masarang 3500 Fuss hoch (Mus. Dresd.), Tomohon (Sarasins).
G r a y hatte zwar die Art von Australien beschrieben, aber
T h o m a s desavouirte diesen Fundort. Ob sie auf Celébes eine
grössere Verbreitung hat, wird die Zukunft lehren.

L y d e k k e r (Intr. Mam. 1891, 477) sagt anmerkungsweise, dass er


Echimys Gray in Echinothrix (PZS. 1867, 59) verbessere, allein
G r a y hat Echiothrix, nicht Echimys. T h o m a s schlug 1896 vor,
Craurothrix für Echinothrix zu gebrauchen, da letzterer Name bereits
vergeben sei.
1 Die Farbe der nackten Theile (Füsse etc.) der Ratten auf dieser, wie den
folgenden 3 Tafeln ist mehr oder weniger nach Gutdünken gewählt, da Angaben
darüber nicht vorliegen. ↑
2 Die goldige Ringelung der einzelnen Haare konnte auf der Abbildung (mit
Handkolorit) nicht wiedergegeben werden. ↑
[Inhalt]
Ungulata
Suidae

[Inhalt]

40. Sus verrucosus celebensis (Müll. Schl.)

Eine a.Kopfhaut mit Schädel eines alten Männchens von


Kalimba am Pik von Bonthain, Süd-Celébes, 26. X 95.
Haut b.eines Weibchens, in Spiritus, aus der Gegend von
Makassar, Süd Celébes.
2 Häute
c, d. mit Schädeln, in Spiritus, von Frischlingen,
l ä n g s g e s t r e i f t , von Kema, Nord-Celébes, XII 94.

Weitere Schädel sind noch in den Händen des Hrn. Dr. S t e h l i n in


Basel, um zusammen mit den Babirusaschädeln der Herren
S a r a s i n (s. unten) bearbeitet zu werden. [28]

Ich weise auf N e h r i n g s Besprechung von Sus celebensis (Abb.


Ber. Dresd. 1888/9 Nr. 2 S. 5–14, Taf. I–II) und bemerke nur, dass
die Sau eine gelbliche Querbinde an der Schnauze und der alte Eber
nur éine Gesichtswarze jederseits besitzt. Im Übrigen scheint mir
F o r s y t h M a j o r ’ s Benennung (AMNH. 6 s. 1897 XIX, 527) die
zweckentsprechendste zu sein. Da das Dresdner Museum
inzwischen ein grösseres Material von Wildschweinen von Nord und
Ost Celébes (im Ganzen jetzt 14 Bälge, 12 Skelette, 4 Schädel, 1
juv. in Spiritus), sowie von den Philippinen erhielt, so hoffe ich darauf
zurückkommen oder das Material Anderen zur Verfügung stellen zu
können.

Die Herren S a r a s i n brachten auch den jungen Schädel mit


Milchgebiss eines Hausschweines von Tomohon, Nord Celébes, mit,
das eventuell vom Wildschwein abstammen könnte.

[Inhalt]

41. Babirusa alfurus Less.

Die Herren S a r a s i n erbeuteten 16 (oder mehr) Babirusa-Schädel


in Celébes, die Hr. Dr. S t e h l i n in Basel zur speciellen Bearbeitung
übernommen hat. Ich benutze aber diese Gelegenheit, indem ich
zugleich auf meine früheren Auseinandersetzungen über Babirusa
alfurus (Abh. Ber. 1896/7 Nr. 6 S. 15–25 Taf. IX) verweise, folgende
Bemerkungen über inzwischen erhaltenes Schädelmaterial zu
machen:

Der (l. c. p. 17) von mir erwähnte angebliche Babirusa-Schädel aus


dem Museum Godeffroy in Hamburg von den Salomo Inseln ist
nunmehr im Leipziger Museum für Völkerkunde aufgefunden
worden, und meine Vermuthung, dass es nur ein Sus-Schädel mit
abnorm gewachsenen unteren Hauern sei, hat sich als richtig
erwiesen (er figurirt im Leipziger Museum jedoch noch als Babirusa-
Schädel). Die oberen Hauer sind frühzeitig entfernt worden, so dass
sich die unteren unbeschränkt entwickeln konnten, allein sie sind in
dieser Entwicklung noch nicht weit vorgeschritten, und die
oberflächliche Ähnlichkeit mit einem Babirusa-Schädel, wenn man
überhaupt von einer solchen reden kann, ist nur im Stand einen
Laien zu täuschen. Übrigens sehe ich nachträglich, dass F i n s c h
dies bereits 1888 (Ethn. Erf. I, 148) richtig gestellt hat. Einen
kreisförmigen Schweinezahn bildete schon E. R o u s s e a u : Anat.
comp. du syst. dent. Paris 1839 T. 20 f. 2 ab.
Hinsichtlich der Frage der o b e r e n E c k z ä h n e b e i d e r
a d u l t e n S a u bemerkte ich l. c. p. 25: „Ob die Normalformel für
den weiblichen Babirusa bezüglich der Eckzähne 0/1 oder 1/1 zu
lauten habe, lässt sich erst sagen, wenn mehr authentische
weibliche Schädel in den Sammlungen sein werden, um zu
erkennen, ob 0/1 oder 1/1 die Ausnahme ist.“ Das Museum erhielt
von der Insel Lembeh bei Nord Celébes einen adulten weiblichen
Schädel (B 3452) von 286 mm Länge, der beiderseits einen mehr
oder weniger horizontal nach vorn und auswärts gerichteten, links
11, rechts 9 mm aus der Alveole hervorragenden, ziemlich spitzen,
oberen Eckzahn hat. Die Alveolarkrämpe (aileron), in der er wurzelt,
ist nicht stärker ausgebildet als bei dem l. c. Taf. IX, 3 von mir
abgebildeten Exemplar ohne oberen Eckzahn. Die Eckzähne des
Unterkiefers ragen links 11, rechts 12 mm aus der Alveole hervor.
Weibliche Schädel sind selten in Museen. Hr. Dr. S t e h l i n theilte
mir mit, dass ihm unter c 80 Schädeln, die er an verschiedenen
Orten gesehen, nur 5 weibliche vorgekommen seien. Sie werden
ihrer Unscheinbarkeit wegen eben an Ort und Stelle nicht
aufbewahrt, während die auffallenden Eckzähne des Männchens
jeden Laien zum Sammeln anregen. Das von mir l. c. p. 24 erwähnte
Leidener semiadulte Exemplar ist nach Dr. S t e h l i n s Ansicht völlig
adult und zeigt auch rechts eine Spur des oberen Eckzahnes in
Form eines Rudimentes; der linke ist zugespitzt. Ein dritter junger
weiblicher Schädel in Leiden, von 177 mm Länge, habe auch keine
Spur eines oberen Eckzahnes, so wenig wie das von mir erwähnte.
Die Herren S a r a s i n hätten aber auch einen alten weiblichen
Schädel mitgebracht mit oberem Eckzahne beiderseits von nicht
ganz 1 cm Länge, ziemlich stumpf, mit einem auf eine sehr mässige
Kante reducirten Alveolarvorsprung.

Ich bin an der Hand dieser Daten jetzt mehr geneigt, c 1/1 für die
Norm und c 0/1 für abnorm anzusehen. Bei seiner Gracilität kann der
Zahn unter Umständen früh ausbrechen oder ist überhaupt deciduös
und sein Fehlen daher, wie in dem von mir l. c. p. 24 beschriebenen
Falle, möglicherweise besser so zu erklären, als durch die Annahme,
dass er nie vorhanden gewesen sei; denn sein Nichtvorhandensein
bei j u n g e n Schädeln mit Milchgebiss oder Resten davon beweist
nicht, dass er nicht schon vorhanden gewesen sein konnte. [29]

In Bezug auf die Z a h n f o r m e l d e s a d u l t e n E b e r s meinte


ich l. c. p. 22, dass es noch sicher gestellt werden müsste, ob in
allen Fällen im definitiven Gebisse 3 Praemolaren auftreten. Das
Dresdner Museum erhielt inzwischen ebenfalls von der Insel
Lembeh bei Nord Celébes einen jungen männlichen Schädel (B
3453) von 249 mm Länge, der in dieser Beziehung lehrreich ist: m 3
überall noch nicht durchgebrochen; im Unterkiefer jederseits 3
Praemolaren, p 3 (der vorderste) aber beiderseits deciduös;
Eckzähne c 22 mm aus der Alveole hervorragend, ihre Wurzeln
reichen aber bereits bis ans hintere Ende von m 2; im Oberkiefer
beiderseits nur 2 Praemolaren, p 3 ist schon ausgefallen, die
Alveolenreste sind jedoch noch vorhanden, und man erkennt hier
deutlich den Grund des Ausfallens: die Wurzeln der Hauer, die c 27
mm aus der Alveole hervorragen, reichen bis an die vordere Wurzel
von p 2 und sind über den alveolaren Löchern der ausgefallenen p 3
sichtbar, sie haben zweifellos das Ausfallen von p 3 mechanisch
bewirkt; p 2 dex. bietet noch die Anomalie, dass er quer steht, die
Längsaxe der Krone ist nicht von vorn nach hinten gerichtet, sondern
von aussen nach innen; der Grund davon liegt zu Tage, indem ein
Praemolar des Milchgebisses zwischen den Alveolen von p 3 und p
2 stehen geblieben ist und noch so fest sitzt, dass man ihn nicht
bewegen kann; p 2 war nicht im Stand ihn hinauszudrängen und hat
sich daher quer stellen müssen. Legt man die Zahnreihen beider
Kiefer aufeinander, so passen sie rechts normal, links aber findet
sich zwischen p 2 und p 1 sup. eine Lücke, da p 2 nicht längs,
sondern quer steht.
Dieser Befund von 3 Praemolaren im Unterkiefer und der sichere
Beweis, dass auch p 3 im Oberkiefer vorhanden gewesen ist, lässt
mich, zusammen mit dem Umstande, dass Hr. Dr. S t e h l i n mir
mittheilt, er habe Spuren von p 3 oder die Zähne selbst öfters
angetroffen, nunmehr annehmen, dass das Vorhandensein von p 3
im Dauergebiss als die Norm zu gelten habe, wenn dieser Zahn
auch meistens früh ausfällt; im Oberkiefer treibt ihn die Wurzel des
Eckzahns mechanisch heraus, im Unterkiefer ist dies bei dem
vorliegenden Schädel (B 3453) nicht der Fall, die Wurzel verläuft im
basalen Theile des Knochens und berührt die Knochen von p 3
nicht.

Endlich bemerke ich über einen auch neuerdings erhaltenen alten


männlichen Schädel (B 3556) von 302 mm Länge aus dem
Gorontaloschen (wo der Babirusa tualangio heisst), dass ihm p 2
sup. sin. fehlt und dass dessen Alveole vollkommen verstrichen ist;
in Folge davon hat sich p 2 inf. sin. abnorm entwickelt, er überragt
mit seiner Spitze die Kaufläche von p 2 um 6 mm, während diese bei
p 2 inf. dex. unter der von p 1 bleibt, und stösst fast an den Rand
des Oberkiefers; p 2 inf. sin. steht mit seiner Basis auch höher als p
1, was wohl ebenfalls eine Folge des fehlenden Antagonisten ist;
denn dass die Wurzel des unteren Hauers die Basis in die Höhe
getrieben haben sollte, ist nicht anzunehmen, weil der
Zwischenraum zwischen ihrer Alveole und dem Kieferrande zu gross
ist. In diesem Fall hat aber auch die Wurzel des oberen Hauers p 2
sup. sin. nicht etwa ausgetrieben, denn ihre Alveole berührt dessen
Basis nicht. Wenn schon, wie wir oben und l. c. p. 22 sahen, p 3
Wechselfällen in höherem Maass ausgesetzt ist, so scheint sich
doch auch p 2 mehr oder weniger, wenn auch seltener, anomal zu
entwickeln, und steht auch dies wohl in Correlation zu dem
aussergewöhnlichen Wachsthume des Eckzahnes.
Bei einem schon länger im Museum aufbewahrten adulten Schädel
von Buru (Nr. 1993), von 284 mm Länge, liegt p 2 sup. sin. nicht
hinter p 2 inf., wie normal, sondern sie stehen übereinander und in
Folge dessen haben sich die Spitzen gegenseitig platt geschliffen.

Was die V e r b r e i t u n g d e s B a b i r u s a anlangt (l. c. p. 15), so


erfuhr ich inzwischen, dass er bei Tolitoli (Nordküste von Celébes)
ganz ausserordentlich häufig vorkomme.
[Inhalt]
Cervidae

[Inhalt]

42. Cervus moluccensis Q. G.

Schädel,
a, b. mas und fem., von Tomohon, Minahassa, Nord
Celébes, III 94.
Schädel,
c. fem., von der Insel Djampea im Süden von
Celébes.
20d–x.
Geweihe: 1 von Kema (Nord Celébes), 3 von
Tomohon (Nord Celébes), 1 aus der Minahassa, 3 von
der Insel Djampea, 12 ohne nähere Bezeichnung aus der
Umgebung des Tominigolfes (in Gorontalo gekauft).

[30]

R ö r i g (Geweihslg. 1896, 49) hat neuerdings den Hirsch von


Celébes nach einem Geweih artlich als „C. celebensis?“ abgetrennt
und (l. c. Fig. 19) abgebildet; er sagt: „Die Geweihform dieser
Species unterscheidet sich insofern von den eben beschriebenen
(equinus), als die von der Hauptstange abgehende Sprosse nicht
hinten oder innen, sondern an der Aussenseite sich abzweigt, so
dass die dadurch entstehende Gabel nicht seitlich, sondern v o r n
offen ist. Die Träger dieser Geweihe bilden in Bezug hierauf somit
den Übergang zu denjenigen Hirschen, bei denen jene Sprosse auf
der Vorderseite der Stange entspringt und auch nach vorn gerichtet
ist, wie wir es z. B. bei den Molukkenhirschen wahrnehmen“ (vgl.
auch seine schematische Tafel zu S. 16). L y d e k k e r (Deer of all
Lands 1898, 166) nennt den Celébeshirsch C. hippelaphus
moluccensis (Q. G.) und nimmt auf R ö r i g keine Rücksicht.
W e b e r (Zool. Erg. I, 112 1890) führte nach Geweihen den
Celébeshirsch als Russa russa S. Müll. ausser von Süd Celébes von
der Insel Saleyer auf, H i c k s o n (Nat. Cel. 1893, 69) von der Insel
Talisse im Norden von Celébes.

Das S a r a s i n sche Material zusammen mit dem des Dresdner


Museums zeigt, dass R ö r i g s Abtrennung der Celébesform von
moluccensis nicht gerechtfertigt ist. Auch ich erbeutete in Süd
Celébes ein grosses Geweih, das die von R ö r i g namhaft
gemachten Charaktere exquisit zeigt, dagegen andere vom Norden
und Süden, die moluccensis entsprechen. Unter den
S a r a s i n schen sind solche, die sich als Übergänge erweisen. Es
ist nicht möglich, die von R ö r i g beschriebene Geweihform als
Altersform anzusehen, da z. B. ein junges Exemplar von
moluccensis, das ich von Ternate mitbrachte, den Charakter bereits
vorzüglich aufweist. Es giebt auch Geweihe, deren eine Stange
mehr zu moluccensis, die andere mehr zu „celebensis“ hinneigt. Das
Geweih dieses Hirsches variirt jedenfalls stark. Ein Fell aber, das
das Dresdner Museum von Nord Celébes besitzt, stimmt sehr gut
mit der Abbildung von Q u o y & G a i m a r d (Voy. Astr. 1833 I pl. 24,
Text 1830 I, 133), die einen Hirsch von Buru darstellt, so dass ich an
der Artzusammengehörigkeit nicht zweifle.

Die Herren S a r a s i n hatten den Eindruck, als ob, nach dem


Geweih zu urtheilen, der nördliche Celébeshirsch grösser sei als der
südliche, das Dresdner grosse Geweih vom Süden bestätigt dies
nicht, allein Endgültiges lässt sich jetzt noch nicht sagen. Wie
G r a a f l a n d (Minahassa 2. Aufl. 1898 App. p. V) mittheilt, wurde
der Hirsch erst Anfang der dreissiger Jahre dieses Jahrhunderts in
die Minahassa eingeführt, die Sprachen dieser Gegend haben daher
auch keine ursprüngliche Bezeichnung für ihn. Man wird ihn wohl
von den Ländern der Tominibucht angebracht haben. Im Süden ist er
sehr häufig, wie ich gelegentlich einer grossen Treibjagd im Jahr

You might also like