3629Hyperbaric and Diving Medicine CHAPTER 463
asthma is considered to increase risk because it could predispose to air
trapping and pulmonary barotrauma (see below); and ischemic heart
disease increases risk because it could prevent a diver from exercising
sufficiently to get out of a difficult situation such as being caught in a
current. It can be a complex matter to recognize the relevant interactions between diving and medical conditions and to determine their
impact on suitability for diving. There may follow an equally complex
discussion about whether such interactions impart a disqualifying
risk, and this may be influenced by the individual candidate’s level of
risk acceptance and the extent to which others involved (such as dive
partners) might be affected. Guidelines are occasionally published
on assessment of diving candidates with risk factors for important
comorbidities like cardiovascular disease or who have suffered topical
problems such as COVID-19 infection (see “Further Reading” list).
Physicians interested in regularly conducting such evaluations should
obtain relevant training. Short courses providing relevant training are
offered by specialist groups in most countries.
BAROTRAUMA
Barotrauma is essentially tissue injury arising as a result of ambient
pressure changes. Middle-ear barotrauma (MEBT) in diving is similar
to the problem that may occur during descent from altitude in an
airplane, but difficulties with equalizing pressure in the middle ear
are exaggerated underwater by both the rapidity and magnitude of
pressure change as a diver descends or ascends. Failure to periodically insufflate the middle-ear spaces via the eustachian tubes during
descent results in increasing pain. As the Pamb increases, the tympanic
membrane (TM) may be bruised or even ruptured as it is pushed
inward. Negative pressure in the middle ear results in engorgement
of blood vessels in the surrounding mucous membranes and leads to
effusion or bleeding, which can be associated with a conductive hearing
loss. MEBT is much less common during ascent because expanding
gas in the middle-ear space tends to open the eustachian tube automatically. Barotrauma may also affect the respiratory sinuses, although
the sinus ostia are usually widely patent and allow automatic pressure
equalization without the need for specific maneuvers. If equalization
fails, pain usually results in termination of the dive. Difficulty with
equalizing ears or sinuses may respond to oral or nasal decongestants.
Much less commonly, divers may suffer inner ear barotrauma
(IEBT). Several explanations have been proposed, of which the most
favored holds that forceful attempts to insufflate the middle-ear space
by Valsalva maneuvers during descent result in transmission of pressure to the perilymph via the cochlear aqueduct and outward rupture
of the round window, which is already under tension because of negative middle-ear pressure. The clinician should be alerted to possible
IEBT after diving by a sensorineural hearing loss or true vertigo (which
is often accompanied by nausea, vomiting, nystagmus, and ataxia).
These manifestations can also occur in vestibulocochlear DCS (see
below) but should never be attributed to MEBT. Immediate review by
an expert diving physician is recommended, and urgent referral to an
otologist will often follow.
The lungs are also vulnerable to barotrauma but are at most risk
during ascent. If expanding gas becomes trapped in the lungs as Pamb
falls, this may rupture alveoli and associated vascular tissue. Gas trapping may occur if divers intentionally or involuntarily hold their breath
during ascent or if there are bullae. The extent to which asthma predisposes to pulmonary barotrauma is debated, but the presence of active
bronchoconstriction must increase risk. For this reason, asthmatics
who regularly require bronchodilator medications or whose airways
are sensitive to exercise or cold air are usually discouraged from diving. While possible consequences of pulmonary barotrauma include
pneumothorax and mediastinal emphysema, the most feared is the
introduction of gas into the pulmonary veins leading to cerebral arterial gas embolism (CAGE). Manifestations of CAGE include loss of
consciousness, confusion, hemiplegia, visual disturbances, and speech
difficulties appearing immediately or within minutes after surfacing.
The management is the same as for DCS described below. The natural
history of CAGE often includes substantial or complete resolution of
symptoms early after the event. This is probably the clinical correlate
of bubble involution and redistribution with consequent restoration
of flow. Patients exhibiting such remissions should still be reviewed
at specialist diving medical centers because secondary deterioration
or re-embolization can occur. Unsurprisingly, these events can be
misdiagnosed as typical strokes or transient ischemic attacks (TIAs)
(Chap. 427) when patients are seen by clinicians unfamiliar with
diving medicine. All patients presenting with neurologic symptoms after
diving should have their symptoms discussed with a specialist in diving
medicine and be considered for recompression therapy.
DECOMPRESSION SICKNESS
DCS is caused by the formation of bubbles from dissolved inert gas
(usually nitrogen) during or after ascent (decompression) from a
compressed gas dive. Bubble formation is also possible following
decompression for extravehicular activity during space flight and with
ascent to altitude in unpressurized aircraft. DCS in the latter scenarios
is probably rare in comparison with diving, where the incidence is
~1:10,000 recreational dives.
Breathing at elevated Pamb results in increased uptake of inert gas
into blood and then into tissues. The rate at which tissue inert gas
equilibrates with the inspired inert gas pressure is proportional to
tissue blood flow and the blood-tissue partition coefficient for the
gas. Similar factors dictate the kinetics of inert gas washout during
ascent. If the rate of gas washout from tissues does not match the rate
of decline in Pamb, then the sum of dissolved gas pressures in the tissue
will exceed Pamb, a condition referred to as “supersaturation.” This is the
prerequisite for bubbles to form during decompression, although other
less well-understood factors are also involved. Deeper and longer dives
result in greater inert gas absorption and greater likelihood of tissue
supersaturation during ascent. Divers control their ascent for a given
depth and time exposure using algorithms that often include periods
where ascent is halted for a prescribed period at different depths to
allow time for gas washout (“decompression stops”). Although a breach
of these protocols increases the risk of DCS, adherence does not guarantee that it will be prevented. DCS should be considered in any diver
manifesting postdive symptoms not readily explained by an alternative
mechanism.
Bubbles may form within tissues themselves, where they cause
symptoms by mechanical distraction of pain-sensitive or functionally
important structures. They also appear in the venous circulation,
almost certainly forming in capillary beds as blood passes through
supersaturated tissues. Some venous bubbles are tolerated without
symptoms and are filtered from the circulation in the pulmonary capillaries. However, in sufficiently large numbers, these bubbles are capable
of inciting inflammatory and coagulation cascades, damaging endothelium, activating formed elements of blood such as platelets, and causing
symptomatic pulmonary vascular obstruction. Moreover, if there is a
right-to-left shunt through a patent foramen ovale (PFO) or an intrapulmonary shunt, then venous bubbles may enter the arterial circulation
(25% of adults have a probe-patent PFO). The risk of cerebral, spinal
cord, inner ear, and skin manifestations appears higher in the presence
of significant shunts, suggesting that these “arterialized” venous bubbles
can cause harm, perhaps by disrupting flow in the microcirculation of
target organs. Circulating microparticles, which are elevated in number
and size after diving, are currently under investigation as indicators of
decompression stress and as injurious agents in their own right. How
they arise and their exact role in DCS remain unclear.
Table 463-3 lists manifestations of DCS grouped according to
organ system. The majority of cases present with mild symptoms,
including musculoskeletal pain, fatigue, and minor neurologic manifestations such as patchy paresthesias. Serious presentations are much
less common. Pulmonary and cardiovascular manifestations can be
life-threatening, and spinal cord involvement frequently results in permanent disability. Latency is variable. Serious DCS usually manifests
within minutes of surfacing, but mild symptoms may not appear for
several hours. Symptoms arising >24 h after diving are very unlikely to
be DCS. The presentation may be confusing and nonspecific, and there
are no useful diagnostic investigations. Diagnosis is based on integration of findings from examination of the dive profile, the nature and
3630 PART 15 Disorders Associated with Environmental Exposures
TABLE 463-3 Manifestations of Decompression Sickness
ORGAN SYSTEM MANIFESTATIONS
Musculoskeletal Limb pain
Neurologic
Cerebral Confusion
Visual disturbances
Speech disturbances
Spinal Muscular weakness
Paralysis
Upper motor neuron signs
Bladder and sphincter dysfunction
Dermatomal sensory disturbances
Abdominal pain
Girdle pain
Vestibulocochlear Hearing loss
Vertigo and ataxia
Nausea and vomiting
Peripheral Patchy nondermatomal sensory disturbance
Pulmonary Cough
Dyspnea
Cardiovascular Hemoconcentration
Coagulopathy
Hypotension
Cutaneous Rash, itch
Lymphatic Soft tissue edema, often relatively localized
Constitutional Fatigue and malaise
■ HYPOTHERMIA
Accidental hypothermia occurs when there is an unintentional drop
in the body’s core temperature below 35°C (95°F). At this temperature,
many of the compensatory physiologic mechanisms that conserve heat
begin to fail. Primary accidental hypothermia is a result of the direct
exposure of a previously healthy individual to the cold. The mortality
rate is much higher for patients who develop secondary hypothermia as
a complication of a serious systemic disorder or injury.
464 Hypothermia and
Peripheral Cold Injuries
Daniel F. Danzl
temporal relationship of symptoms, and the clinical examination. Some
DCS presentations may be difficult to separate from CAGE following
pulmonary barotrauma, but from a clinical perspective, the distinction
is unimportant because the first aid and definitive management of both
conditions are the same.
TREATMENT
Diving Medicine
First aid for either DCS or CAGE includes horizontal positioning
(especially if there are cerebral manifestations), intravenous fluids
if available, and sustained 100% oxygen administration. The latter
accelerates inert gas washout from tissues and promotes resolution of
bubbles. Definitive treatment of DCS or CAGE with recompression
and hyperbaric oxygen is justified in most instances, although some
mild or marginal DCS cases may be managed with first aid measures alone—an option that may be invoked by experienced diving
physicians under various circumstances, but especially if evacuation
for recompression is hazardous or extremely difficult. Long-distance
evacuations are usually undertaken using a helicopter flying at low
altitude or a fixed-wing air ambulance pressurized to 1 ATA.
Recompression reduces bubble volume in accordance with
Boyle’s law and increases the inert gas partial pressure difference
between a bubble and surrounding tissue. At the same time,
oxygen administration markedly increases the inert gas partial
pressure difference between alveoli and tissue. The net effect is to
significantly increase the rate of inert gas diffusion from bubble
to tissue and tissue to blood, thus accelerating bubble resolution.
Hyperbaric oxygen also helps oxygenate compromised tissues and
may ameliorate some of the proinflammatory effects of bubbles.
Various recompression protocols have been advocated, but there
are no data that define the optimum approach. Recompression typically begins with oxygen administered at 2.8 ATA, the maximum
pressure at which the risk of oxygen toxicity remains acceptable in
a hyperbaric chamber. There follows a stepwise decompression over
variable periods adjusted to symptom response. The most widely
used algorithm is the U.S. Navy Table 6, whose shortest format lasts
4 h and 45 min. Typically, shorter “follow-up” recompressions are
repeated daily while symptoms persist and appear responsive to
treatment. Adjuncts to recompression include intravenous fluids
and other supportive care as necessary. Occasionally, very sick
divers require intubation, ventilation, and high-level intensive care.
The presentation of sick divers to physicians or hospitals without
diving medicine expertise creates a risk of misinterpretation of
nonspecific manifestations and of consequent mistakes in diagnosis and management. Physicians finding themselves in this situation are strongly advised to expeditiously contact the 24-h diving
emergency advisory service provided by the Divers Alert Network
(DAN). This can be accessed at +1-919-684-9111, and there are
subsidiary or related services in virtually all jurisdictions globally.
■ FURTHER READING
Bennett MH et al: Hyperbaric oxygen therapy for late radiation tissue
injury. Cochrane Database Syst Rev 4:CD005005, 2016.
Edmonds C et al: Diving and Subaquatic Medicine, 5th ed. Boca Raton,
FL, Taylor and Francis, 2015.
Francis A, Baynosa R: Ischaemia-reperfusion injury and hyperbaric
oxygen pathways: A review of cellular mechanisms. Diving Hyperb
Med 47:110, 2017.
Gorenstein SA et al: Hyperbaric oxygen therapy for COVID-19
patients with respiratory distress: Treated cases versus propensity
-matched controls. Undersea Hyperb Med 47:405, 2020.
Jepson N et al: South Pacific Underwater Medicine Society guidelines
for cardiovascular risk assessment of divers. Diving Hyperb Med
50:273, 2020.
Kranke P et al: Hyperbaric oxygen therapy for chronic wounds.
Cochrane Database Syst Rev 6:CD004123, 2015.
Mitchell SJ et al: Pre-hospital management of decompression illness:
Expert review of key principles and controversies. Diving Hyperb
Med 48:45, 2018.
Moon RE (ed): Hyperbaric Oxygen Therapy Indications, 14th ed. North
Palm Beach, FL, Best Publishing Company, 2018.
Oley MH et al: Effects of hyperbaric oxygen therapy on vascular endothelial growth factor protein and mRNA in crush injury patients: A
randomized controlled trial study. Int J Surg Open 29:33, 2021.
Sadler C et al: Diving after SARS-CoV-2 (COVID-19) infection: Fitness to dive assessment and medical guidance. Diving Hyperb Med
50:278, 2020.
Vann RD et al: Decompression illness. Lancet 377:153, 2011.
Weaver LK et al: Hyperbaric oxygen for acute carbon monoxide poisoning. N Engl J Med 347:1057, 2002.
Whelan HT, Kindwall EP (eds): Hyperbaric Medicine Practice,
4th ed. Palm Beach, FL, Best Publishing Company, 2017.
3631Hypothermia and Peripheral Cold Injuries CHAPTER 464
disrupts the autonomic pathways that lead to shivering and will prevent
cold-induced reflex vasoconstrictive responses.
Hypothermia associated with sepsis is a poor prognostic sign.
Hepatic failure causes decreased glycogen storage and gluconeogenesis as well as a diminished shivering response. In acute myocardial
infarction associated with low cardiac output, hypothermia may be
reversed after adequate resuscitation. With extensive burns, psoriasis,
erythrodermas, and other skin diseases, increased peripheral-blood
flow leads to excessive heat loss.
■ THERMOREGULATION
Heat loss occurs through five mechanisms: radiation (55–65% of heat
loss), conduction (10–15% of heat loss, increased in cold water), convection (increased in the wind), respiration, and evaporation; both of
the latter two mechanisms are affected by the ambient temperature and
the relative humidity.
The preoptic anterior hypothalamus normally orchestrates thermoregulation (Chap. 18). The immediate defense of thermoneutrality
is via the autonomic nervous system, whereas delayed control is mediated by the endocrine system. Autonomic nervous system responses
include the release of norepinephrine, increased muscle tone, and shivering, leading to thermogenesis and an increase in the basal metabolic
rate. Cutaneous cold thermoreception causes direct reflex vasoconstriction to conserve heat. Prolonged exposure to cold also stimulates
the thyroid axis, leading to an increased metabolic rate.
■ CLINICAL PRESENTATION
In most cases of hypothermia, the history of exposure to environmental factors (e.g., prolonged exposure to the outdoors without adequate
clothing) makes the diagnosis straightforward. In urban settings,
however, the presentation is often more subtle, and other disease processes, toxin exposures, or psychiatric diagnoses should be considered.
Predicting the core temperature based on the clinical presentation is
very difficult.
After initial stimulation by hypothermia, there is progressive
depression of all organ systems. The timing of the appearance of these
clinical manifestations varies widely (Table 464-2). Without knowing the core temperature, it can be difficult to interpret other vital
signs. For example, tachycardia disproportionate to the core temperature suggests secondary hypothermia resulting from hypoglycemia,
hypovolemia, or a toxin overdose. Because carbon dioxide production
declines progressively, the respiratory rate should be low; persistent
hyperventilation suggests a central nervous system (CNS) lesion or
an organic acidosis. A markedly depressed level of consciousness in a
patient with mild hypothermia suggests an overdose or CNS dysfunction due to infection or trauma.
Physical examination findings will also be altered by hypothermia.
For instance, the assumption that areflexia is solely attributable to
hypothermia can obscure the diagnosis of a spinal cord lesion. Patients
with hypothermia may be confused or combative; these symptoms
abate more rapidly with rewarming than with chemical or physical
restraint. A classic example of maladaptive behavior in patients with
hypothermia is paradoxical undressing, which involves the inappropriate removal of clothing in response to a cold stress. The cold-induced
ileus and abdominal rectus spasm can mimic or mask the presentation
of an acute abdomen (Chap. 15).
When a patient in hypothermic cardiac arrest is first discovered,
cardiopulmonary resuscitation (CPR) is indicated unless (1) a do-notresuscitate status is verified, (2) obviously lethal injuries are identified,
or (3) the depression of a frozen chest wall is not possible. Continuous
CPR is normally recommended, and interruptions should be avoided if
possible. In the field, when the core temperature is <28°C, intermittent
CPR may also be effective.
As the resuscitation proceeds, the prognosis is grave if there is evidence of widespread cell lysis, as reflected by potassium levels >10–12
mmol/L (10–12 meq/L). Other findings that may preclude continuing
resuscitation include a core temperature <10–12°C (<50–54°F), a
pH <6.5, and evidence of intravascular thrombosis with a fibrinogen
value <0.5 g/L (<50 mg/dL). The decision to terminate resuscitation
TABLE 464-1 Risk Factors for Hypothermia
Age extremes
Elderly
Neonates
Environmental exposure
Occupational
Sports-related
Inadequate clothing
Immersion
Toxicologic and pharmacologic
Ethanol
Anesthetics
Antipsychotics
Antidepressants
Anxiolytics
Benzodiazepines
Neuromuscular blockers
Insufficient fuel
Malnutrition
Marasmus
Kwashiorkor
Endocrine-related
Diabetes mellitus
Hypoglycemia
Hypothyroidism
Adrenal insufficiency
Hypopituitarism
Neurologic
Cerebrovascular accident
Hypothalamic disorders
Parkinson’s disease
Spinal cord injury
Multisystemic
Trauma
Sepsis
Shock
Hepatic or renal failure
Carcinomatosis
Burns and exfoliative dermatologic
disorders
Immobility or debilitation
■ CAUSES
Primary accidental hypothermia is geographically and seasonally pervasive. Although most cases occur in the winter months and in colder
climates, this condition is surprisingly common in warmer regions as
well. Multiple variables render individuals at the extremes of age—both
the elderly and neonates—particularly vulnerable to hypothermia
(Table 464-1). The elderly have diminished thermal perception and are
more susceptible to immobility, malnutrition, and systemic illnesses
that interfere with heat generation or conservation. Dementia, psychiatric illness, and socioeconomic factors often compound these problems. Neonates have high rates of heat loss because of their increased
surface-to-mass ratio and their lack of effective shivering and adaptive
behavioral responses. At all ages, malnutrition can contribute to heat
loss because of diminished subcutaneous fat and as a result of depleted
energy stores used for thermogenesis.
Individuals whose occupations or hobbies entail extensive exposure
to cold weather are at increased risk for hypothermia. Military history
is replete with hypothermic tragedies. Hunters, sailors, skiers, and
climbers also are at great risk of exposure, whether it involves injury,
changes in weather, or lack of preparedness.
Ethanol causes vasodilation (which increases heat loss), reduces
thermogenesis and gluconeogenesis, and may impair judgment or lead
to obtundation. Some antipsychotics, antidepressants, anxiolytics, benzodiazepines, and other medications reduce centrally mediated vasoconstriction. Many hypothermic patients are admitted to intensive care
because of drug overdose. Anesthetics can block shivering responses;
these effects are compounded when patients are not insulated adequately in the operating or recovery units.
Several types of endocrine dysfunction cause hypothermia.
Hypothyroidism—particularly when extreme, as in myxedema coma—
reduces the metabolic rate and impairs thermogenesis and behavioral
responses. Adrenal insufficiency and hypopituitarism also increase
susceptibility to hypothermia. Hypoglycemia, most commonly caused
by insulin or oral hypoglycemic agents, is associated with hypothermia,
in part because of neuroglycopenic effects on hypothalamic function.
Increased osmolality and metabolic derangements associated with
uremia, diabetic ketoacidosis, and lactic acidosis can lead to altered
hypothalamic thermoregulation.
Neurologic injury from trauma, cerebrovascular accident, subarachnoid hemorrhage, and a hypothalamic lesion increases susceptibility to
hypothermia. Agenesis of the corpus callosum (Shapiro’s syndrome) is
one cause of episodic hypothermia. In this syndrome, profuse perspiration is followed by a rapid fall in temperature. Acute spinal cord injury
3632 PART 15 Disorders Associated with Environmental Exposures
before rewarming the patient past 33°C (91°F) should be predicated
on the type and severity of the precipitants of hypothermia. Survival
has occurred with a cardiac arrest time over 7 h. There is an ongoing
search for validated prognostic indicators for recovery from hypothermia. The Swiss grading system considers core body temperature
and the clinical findings. Other scoring systems also consider age,
albumin, and lactate levels. A history of asphyxia, as in an avalanche,
with secondary cooling is the most important negative predictor of
survival.
■ DIAGNOSIS AND STABILIZATION
Hypothermia is confirmed by measurement of the core temperature,
preferably at two sites. Rectal probes should be placed to a depth of
15 cm and not adjacent to cold feces. A simultaneous esophageal probe
can be placed 24 cm below the larynx; it may read falsely high during
heated inhalation therapy. Relying solely on infrared tympanic thermography is not advisable.
After a diagnosis of hypothermia is established, cardiac monitoring
should be instituted, along with attempts to limit further heat loss. If
the patient is in ventricular fibrillation, it is unclear at what core temperature ventricular defibrillation (2 J/kg) should first be attempted.
One biphasic attempt below 30°C is warranted. Further defibrillation
attempts should usually be deferred until some rewarming (1°–2°C)
is achieved and ventricular fibrillation is coarser. Although cardiac
pacing for hypothermic bradydysrhythmias is rarely indicated, the
transthoracic technique is preferable. The J or Osborn wave at the
junction of the QRS complex and ST segment suggests the diagnosis.
Obvious J waves are routinely misdiagnosed by automated readings as
injury current.
Supplemental oxygenation is always warranted, since tissue oxygenation is affected adversely by the leftward shift of the oxyhemoglobin
dissociation curve. Pulse oximetry is often unreliable in patients with
vasoconstriction. If protective airway reflexes are absent, gentle endotracheal intubation should be performed. Adequate preoxygenation
will prevent ventricular arrhythmias.
Insertion of a gastric tube prevents dilation secondary to decreased
bowel motility. Indwelling bladder catheters facilitate monitoring
of cold-induced diuresis and can provide an ancillary approach for
temperature monitoring. Dehydration is encountered commonly with
chronic hypothermia, and most patients benefit from an intravenous
or intraosseous crystalloid bolus. Normal saline is preferable to lactated
Ringer’s solution, as the liver in hypothermic patients inefficiently
metabolizes lactate. The placement of a pulmonary artery catheter can
cause perforation of the less compliant pulmonary artery. Insertion of
a central venous catheter deeply into the cold right atrium should be
avoided since this procedure, similar to transvenous pacing, can precipitate refractory arrhythmias.
Arterial blood gases should not be corrected for temperature
(Chap. 55). An uncorrected pH of 7.42 and a Pco2
of 40 mmHg reflect
appropriate alveolar ventilation and acid-base balance at any core temperature. Acid-base imbalances should be corrected gradually, since
the bicarbonate buffering system is inefficient. A common error is
overzealous hyperventilation in the setting of depressed CO2
production. When the Pco2
decreases by 10 mmHg at 28°C (82°F), it doubles
the pH increase of 0.08 that occurs at 37°C (99°F).
The severity of anemia may be underestimated because the hematocrit increases 2% for each 1°C drop in temperature. White blood cell
sequestration and bone marrow suppression are common, potentially
masking an infection. Although hypokalemia is more common in
chronic hypothermia, hyperkalemia also occurs; the expected electrocardiographic changes are often obscured by hypothermia. Patients
with renal insufficiency, metabolic acidosis, or rhabdomyolysis are at
greatest risk for electrolyte disturbances.
Coagulopathies are common because cold inhibits the enzymatic
reactions required for activation of the intrinsic cascade. In addition,
thromboxane B2
production by platelets is temperature dependent,
and platelet function is impaired. The administration of platelets and
fresh-frozen plasma is therefore not effective. Coagulation studies can
be deceptively normal and contrast with the observed in vivo coagulopathy. This contradiction occurs because all coagulation tests are routinely performed at 37°C (99°F), and the enzymes are thus rewarmed.
■ REWARMING STRATEGIES
The key initial decision is whether to rewarm the patient passively or
actively. Passive external rewarming simply involves covering and insulating the patient in a warm environment. With the head also covered,
the rate of rewarming is usually 0.5°–2°C (1.10°–4.4°F) per hour. This
technique is ideal for previously healthy patients who develop acute,
mild primary accidental hypothermia. The patient must have sufficient
glycogen to support endogenous thermogenesis.
The application of heat directly to the extremities of patients with
chronic severe hypothermia should be avoided because it can induce
peripheral vasodilation and precipitate core temperature “afterdrop,” a
response characterized by a continual decline in the core temperature
TABLE 464-2 Physiologic Changes Associated with Accidental Hypothermia
SEVERITY BODY TEMPERATURE
CENTRAL NERVOUS
SYSTEM CARDIOVASCULAR RESPIRATORY
RENAL AND
ENDOCRINE NEUROMUSCULAR
Mild 35°C (95°F)–32.2°C
(90°F)
Linear depression of
cerebral metabolism;
amnesia; apathy;
dysarthria; impaired
judgment; maladaptive
behavior
Tachycardia,
then progressive
bradycardia; cardiac
cycle prolongation;
vasoconstriction;
increase in cardiac output
and blood pressure
Tachypnea, then
progressive decrease
in respiratory minute
volume; declining
oxygen consumption;
bronchorrhea;
bronchospasm
Diuresis; increase
in catecholamines,
adrenal steroids,
triiodothyronine, and
thyroxine; increase
in metabolism with
shivering
Increased
preshivering muscle
tone, then fatiguing
Moderate <32.2°C (90°F)–28°C
(82.4°F)
EEG abnormalities;
progressive depression
of level of consciousness;
pupillary dilation;
paradoxical undressing;
hallucinations
Progressive decrease in
pulse and cardiac output;
increased atrial and
ventricular arrhythmias;
suggestive (J-wave) ECG
changes
Hypoventilation: 50%
decrease in carbon
dioxide production
per 8°C (17.6°F)
drop in temperature;
absence of protective
airway reflexes
50% increase in renal
blood flow; renal
autoregulation intact;
impaired insulin
action
Hyporeflexia;
diminishing
shivering-induced
thermogenesis; rigidity
Severe <28°C (<82.4°F) Loss of cerebrovascular
autoregulation; decline
in cerebral blood flow;
coma; loss of ocular
reflexes; progressive
decrease in EEG
abnormalities
Progressive decrease
in blood pressure, heart
rate, and cardiac output;
reentrant dysrhythmias;
maximal risk of ventricular
fibrillation; asystole
Pulmonic congestion
and edema; 75%
decrease in oxygen
consumption; apnea
Decrease in renal
blood flow that
parallels decrease
in cardiac output;
extreme oliguria;
poikilothermia; 80%
decrease in basal
metabolism
No motion; decreased
nerve-conduction
velocity; peripheral
areflexia; no corneal
or oculocephalic
reflexes
Abbreviations: ECG, electrocardiogram; EEG, electroencephalogram.
Source: From DF Danzl, RS Pozos: Accidental hypothermia. N Engl J Med 331:1756, 1994. Copyright © 1994 Massachusetts Medical Society. Reprinted with permission from
Massachusetts Medical Society.
3633Hypothermia and Peripheral Cold Injuries CHAPTER 464
after removal of the patient from the cold. Truncal heat application
reduces the risk of afterdrop.
Active rewarming is necessary under the following circumstances:
core temperature <32°C (<90°F) (poikilothermia), cardiovascular instability, age extremes, CNS dysfunction, hormone insufficiency, and
suspicion of secondary hypothermia. Active external rewarming is best
accomplished with forced-air heating blankets. Other options include
devices that circulate water through external heat exchange pads,
radiant heat sources, and hot packs. Monitoring a patient with hypothermia in a heated tub is extremely difficult. Electric blankets should
be avoided because vasoconstricted skin is easily burned.
There are numerous widely available options for active core
rewarming. Airway rewarming with heated humidified oxygen (40°–
45°C [104°–113°F]) via mask or endotracheal tube is a convenient
option. Although airway rewarming provides less heat than do some
other forms of active core rewarming, it eliminates respiratory heat
loss and adds 1°–2°C (2.2°–4.4°F) to the overall rewarming rate. Crystalloids should be heated to 40°–42°C (104°–108°F), but the quantity of
heat provided is significant only during massive volume resuscitation.
The most efficient method for heating and delivering fluid or blood is
with a countercurrent in-line heat exchanger. Heated irrigation of the
gastrointestinal tract or bladder transfers minimal heat because of the
limited available surface area. These methods should be reserved for
patients in cardiac arrest and then used in combination with all available active rewarming techniques.
Closed thoracic lavage is far more efficient in severely hypothermic
patients with cardiac arrest. The hemithoraxes are irrigated through
two inserted large-bore thoracostomy tubes. Thoracostomy tubes
should not be placed in the left chest of a spontaneously perfusing
patient for purposes of rewarming. Peritoneal lavage with the dialysate
at 40°–45°C (104°–113°F) efficiently transfers heat when delivered
through two catheters with outflow suction. Like peritoneal dialysis,
standard hemodialysis is especially useful for patients with electrolyte
abnormalities, rhabdomyolysis, or toxin ingestion. Another option
involves the use of endovascular temperature control catheters.
Extracorporeal blood rewarming options (Table 464-3) should be
considered in severely hypothermic patients, especially those with
primary accidental hypothermia. Extracorporeal life support, including
bypass, should be considered in nonperfusing patients without documented contraindications to resuscitation. Circulatory support may be
the only effective option in patients with completely frozen extremities
or those with significant tissue destruction coupled with rhabdomyolysis. There is no evidence that extremely rapid rewarming improves
survival in perfusing patients.
TREATMENT
Hypothermia
When a patient is hypothermic, target organs and the cardiovascular system respond minimally to most medications. Generally,
medications are withheld below 30°C (86°F). In contrast to antiarrhythmics, low-dose vasopressor medications may improve the
intra-arrest rates of return of spontaneous circulation. Because of
increased binding of drugs to proteins as well as impaired metabolism and excretion, either a lower dose or a longer interval between
doses should be used to avoid toxicity. As an example, the administration of repeated doses of digoxin or insulin would be ineffective
while the patient is hypothermic, but the residual drugs would be
potentially toxic during rewarming.
Achieving a mean arterial pressure of at least 60 mmHg should
be an early objective. If the hypotension is disproportionate for
temperature and does not respond to crystalloid/colloid infusion
and rewarming, low-dose dopamine support (2–5 μg/kg per min)
should be considered. Perfusion of the vasoconstricted cardiovascular system also may improve with low-dose IV nitroglycerin.
Atrial arrhythmias should be monitored initially without intervention, as the ventricular response should be slow and, unless
preexistent, most will convert spontaneously during rewarming.
The role of prophylaxis and treatment of ventricular arrhythmias
is complex. Preexisting ventricular ectopy may be suppressed by
hypothermia and reappear during rewarming. None of the class I
agents is proven to be safe and efficacious.
Initiating empirical therapy for adrenal insufficiency usually is
not warranted unless the history suggests steroid dependence or
hypoadrenalism or efforts to rewarm with standard therapy fail. The
administration of parenteral levothyroxine to euthyroid patients
with hypothermia, however, is potentially hazardous. Because laboratory results can be delayed and confounded by the presence of
the sick euthyroid syndrome (Chap. 382), historic clues or physical
findings suggestive of hypothyroidism should be sought. When
myxedema is the cause of hypothermia, the relaxation phase of the
Achilles reflex is prolonged more than is the contraction phase.
Hypothermia obscures most of the symptoms and signs of infection, notably fever and leukocytosis. Shaking rigors from infection
may be mistaken for shivering. Except in mild cases, extensive
cultures and repeated physical examinations are essential. Unless
an infectious source is identified, empirical antibiotic prophylaxis is
most warranted in the elderly, neonates, and immunocompromised
patients.
FROSTBITE
Peripheral cold injuries include both freezing and nonfreezing injuries
to tissue. Tissue freezes quickly when in contact with thermal conductors such as metal and volatile solutions. Other predisposing factors
include constrictive clothing or boots, immobility, and vasoconstrictive
medications. Frostbite occurs when the tissue temperature drops below
0°C (32°F). Ice-crystal formation subsequently distorts and destroys
the cellular architecture. Once the vascular endothelium is damaged,
stasis progresses rapidly to microvascular thrombosis. After the tissue
thaws, there is progressive dermal ischemia. The microvasculature
begins to collapse, arteriovenous shunting increases tissue pressures,
and edema forms. Finally, thrombosis, ischemia, and superficial necrosis appear. The development of mummification and demarcation may
take weeks to months.
TABLE 464-3 Options for Extracorporeal Blood Rewarming
EXTRACORPOREAL
REWARMING TECHNIQUE CONSIDERATIONS
Continuous venovenous (CVV)
rewarming
Circuit: CV catheter to CV, dual-lumen CV, or
peripheral catheter
No oxygenator/circulatory support
Flow rates 150–400 mL/min
ROR 2°–3°C (4.4°–6.6°F)/h
Hemodialysis Circuit: single- or dual-vessel cannulation
Stabilizes electrolyte or toxicologic
abnormalities
Exchange cycle volumes 200–500 mL/min
ROR 2°–3°C (4.4°–6.6°F)/h
Continuous arteriovenous
rewarming (CAVR)
Circuit: percutaneous 8.5-Fr femoral catheters
Requires systolic blood pressure of 60 mmHg
No perfusionist/pump/anticoagulation
Flow rates 225–375 mL/min
ROR 3°–4°C (6.6°–8.8°F)/h
Cardiopulmonary bypass (CPB) Circuit: full circulatory support with pump and
oxygenator
Perfusate-temperature gradient 5°–10°C
(11°–22° F)
Flow rates 2–7 L/min (average 3–4 L/min)
ROR up to 9.5°C (20.9°F)/h
Venoarterial extracorporeal
membrane oxygenation
(VA-ECMO)
Decreased risk of post-rewarming
cardiorespiratory failure
Improved neurologic outcome
Abbreviations: CV, central venous; ROR, rate of rewarming.
3634 PART 15 Disorders Associated with Environmental Exposures
CLINICAL PRESENTATION
The initial presentation of frostbite can be deceptively benign. The
symptoms always include a sensory deficiency affecting light touch,
pain, or temperature perception. The acral areas and distal extremities
are the most common insensate areas. Some patients describe a clumsy
or “chunk of wood” sensation in the extremity.
Deep frostbitten tissue can appear waxy, mottled, yellow, or
violaceous-white. Favorable presenting signs include some warmth
or sensation with normal color. The injury is often superficial if the
subcutaneous tissue is pliable or if the dermis can be rolled over bony
prominences.
Clinically, frostbite is superficial or deep. Superficial frostbite does
not entail tissue loss but rather causes only anesthesia and erythema.
The appearance of vesiculation surrounded by edema and erythema
implies deeper involvement (Fig. 464-1). Hemorrhagic vesicles reflect
a serious injury to the microvasculature and indicate severe frostbite.
Damages in subcuticular, muscular, or osseous tissues may result in
amputation. An alternative classification establishes grades based on
the location of presenting cyanosis; that is grade 1, absence of cyanosis;
grade 2, cyanosis on the distal phalanx; grade 3, cyanosis up to the
metacarpophalangeal (MP) joint; and grade 4 cyanosis proximal to the
MP joint.
The two most common nonfreezing peripheral cold injuries are
chilblain (pernio) and immersion (trench) foot. Chilblain results from
neuronal and endothelial damage induced by repetitive exposure to
damp cold above the freezing point. Young females, particularly those
with a history of Raynaud’s phenomenon, are at greatest risk. Persistent
vasospasticity and vasculitis can cause erythema, mild edema, and pruritus. Eventually plaques, blue nodules, and ulcerations develop. These
TABLE 464-4 Treatment for Frostbite
BEFORE THAWING DURING THAWING AFTER THAWING
Remove from
environment.
Consider parenteral
analgesia and ketorolac.
Gently dry and protect part;
elevate; place pledgets
between toes, if macerated.
Prevent partial
thawing and
refreezing.
Administer ibuprofen
(400 mg PO).
If clear vesicles are intact,
aspirate sterilely; if broken,
debride and dress with
antibiotic or sterile aloe vera
ointment.
Stabilize core
temperature and treat
hypothermia.
Immerse part in
37°–40°C (99°–104°F)
(thermometer-monitored)
circulating water
containing an antiseptic
soap until distal flush
(10–45 min).
Leave hemorrhagic vesicles
intact to prevent desiccation
and infection.
Protect frozen
part—no friction or
massage.
Encourage patient to
gently move part.
Continue ibuprofen
(400–600 mg PO [12 mg/kg
per day] q8 to 12h).
Address medical or
surgical conditions.
If pain is refractory,
reduce water
temperature to 35°–37°C
(95°–99°F) and administer
parenteral narcotics.
Consider tetanus and
streptococcal prophylaxis;
elevate part.
Administer hydrotherapy at
37°C (99°F).
Consider dextran or
phenoxybenzamine or, in
severe cases, thrombolysis
rt-PA (IV or intraarterial).
FIGURE 464-1 Frostbite with vesiculation, surrounded by edema and erythema. Abbreviation: rt-PA, recombinant tissue plasminogen activator.
lesions typically involve the dorsa of the hands and feet. In contrast,
immersion foot results from repetitive exposure to wet cold above the
freezing point. The feet initially appear cyanotic, cold, and edematous.
The subsequent development of bullae is often indistinguishable from
frostbite. This vesiculation rapidly progresses to ulceration and liquefaction gangrene. Patients with milder cases report hyperhidrosis, cold
sensitivity, and painful ambulation for many years.
TREATMENT
Peripheral Cold Injuries
When frostbite accompanies hypothermia, hydration may improve
vascular stasis. Frozen tissue should be thawed rapidly and completely by immersion in circulating water at 37°–40°C (99°–104°F)
for 30–60 min and not by using hot air. Rapid rewarming often
produces an initial hyperemia. The early formation of large clear
distal blebs is more favorable than that of smaller proximal dark
hemorrhagic blebs. A common error is the premature termination
of thawing, since the reestablishment of perfusion is intensely painful. Parenteral narcotics will be necessary with deep frostbite. If
cyanosis persists after rewarming, the tissue compartment pressures
should be monitored carefully.
Many antithrombotic and vasodilatory treatment regimens have
been evaluated. The prostacyclin analogue iloprost given within
48 h after rewarming is an option. There is no conclusive evidence
that sympathectomy, steroids, calcium channel blockers, or hyperbaric oxygen salvages tissue.
Patients who have deep frostbite injuries with the potential for
significant morbidity should be considered for intravenous or
intraarterial thrombolytic therapy. Angiography or pyrophosphate
scanning may help evaluate the injury and monitor the progress
of tissue plasminogen activator therapy (rt-PA). Heparin is recommended as adjunctive therapy. Intraarterial thrombolysis may
reduce the need for digital and more proximal amputations when
administered within 24 h of severe injuries. A treatment protocol
for frostbite is summarized in Table 464-4.
Unless infection develops, any decision regarding debridement or amputation should generally be deferred. Angiography or
3635Heat-Related Illnesses CHAPTER 465
technetium-99 bone scan may assist in the determination of surgical margins. Magnetic resonance angiography may also demonstrate the line of demarcation earlier than does clinical demarcation.
The most common symptomatic sequelae reflect neuronal injury
and persistently abnormal sympathetic tone, including paresthesia, thermal misperception, and hyperhidrosis. Delayed findings
include nail deformities, cutaneous carcinomas, and epiphyseal
damage in children.
Management of the chilblain syndrome is usually supportive.
With refractory perniosis, alternatives include nifedipine, steroids,
and limaprost, a prostaglandin E1
analogue.
■ FURTHER READING
Cauchy E et al: A controlled trial of a prostacyclin and rt-PA in the
treatment of severe frostbite. N Engl J Med 364:189, 2011.
McIntosh SE et al: Wilderness Medical Society practice guidelines for
the prevention and treatment of frostbite. Wilderness Environ Med
25(4 suppl):S43, 2014.
Ohbe H et al: Extracorporeal membrane oxygenation improves outcomes of accidental hypothermia without vital signs: A nationwide
observational study. Resuscitation 144:27, 2019.
Okada Y et al: The development and validation of a “5A” severity scale
for predicting in-hospital mortality after accidental hypothermia
from J-point registry data. J Intensive Care 7:27, 2019.
Zafren K: Out-of-hospital evaluation and treatment of accidental
hypothermia. Emerg Med Clin North Am 35:261, 2017.
Heat-related illnesses include a spectrum of disorders ranging from
heat syncope, muscle cramps, and heat exhaustion to medical emergencies such as heatstroke. The core body temperature is normally
maintained within a very narrow range. Although significant levels of
hypothermia are tolerated (Chap. 464), multiorgan dysfunction occurs
rapidly at temperatures >41°–43°C. In contrast to heatstroke, the far
more common sign of fever reflects intact thermoregulation.
■ THERMOREGULATION
Humans are capable of significant heat generation. Strenuous exercise
can increase heat generation twentyfold. The heat load from metabolic
heat production and environmental heat absorption is balanced by a
variety of heat dissipation mechanisms. These central integrative dissipation pathways are orchestrated by the central thermostat, which is
located in the preoptic nucleus of the anterior hypothalamus. Efferent
signals sent via the autonomic nervous system trigger cutaneous vasodilation and diaphoresis to facilitate heat loss.
Normally, the body dissipates heat into the environment via four
mechanisms. The evaporation of skin moisture is the single most efficient mechanism of heat loss but becomes progressively ineffective as
the relative humidity rises to >70%. The radiation of infrared electromagnetic energy directly into the surrounding environment occurs
continuously. (Conversely, radiation is a major source of heat gain in hot
climates.) Conduction—the direct transfer of heat to a cooler object—
and convection—the loss of heat to air currents—become ineffective
when the environmental temperature exceeds the skin temperature.
Factors that interfere with the evaporation of diaphoresis significantly
increase the risk of heat illness. Examples include dripping of sweat off
the skin, constrictive or occlusive clothing, dehydration, and excessive
humidity. While air is an effective insulator, the thermal conductivity
of water is 25 times greater than that of air at the same temperature.
465 Heat-Related Illnesses
Daniel F. Danzl
The wet-bulb globe temperature is a commonly used index to assess
the environmental heat load. This calculation considers the ambient
air temperature, the relative humidity, and the degree of radiant heat.
The regulation of this heat load is complex and involves the central nervous system (CNS), thermosensors, and thermoregulatory
effectors. The central thermostat activates the effectors that produce
peripheral vasodilation and sweating. The skin surface is in effect the
radiator and the principal location of heat loss, since skin blood flow
can increase 25–30 times over the basal rate. This dramatic increase in
skin blood flow, coupled with the maintenance of peripheral vasodilation, efficiently radiates heat. At the same time, there is a compensatory
vasoconstriction of the splanchnic and renal beds.
Acclimatization to heat reflects a constellation of physiologic adaptations that permit the body to lose heat more efficiently. This process
often requires one to several weeks of exposure and work in a hot
environment. During acclimatization, the thermoregulatory set point
is altered, and this alteration affects the onset, volume, and content
of diaphoresis. The threshold for the initiation of sweating is lowered,
and the amount of sweat increases, with a lowered salt concentration.
Sweating rates can be 1–2 L/h in acclimated individuals during heat
stress. Plasma volume expansion also occurs and improves cutaneous
vascular flow. The heart rate lowers, with a higher stroke volume.
After the individual leaves the hot environment, improved tolerance
to heat stress dissipates rapidly, the plasma volume decreases, and deacclimatization occurs within weeks.
■ PREDISPOSING FACTORS AND
DIFFERENTIAL DIAGNOSIS
When there is an excessive heat load, unacclimated individuals can
develop a variety of heat-related illnesses. Heat waves exacerbate the
mortality rate, particularly among the elderly and among persons
lacking adequate nutrition and access to air-conditioned environments.
Secondary vascular events, including cerebrovascular accidents and
myocardial infarctions, occur at least 10 times more often in conditions
of extreme heat.
Exertional heat illness continues to occur when laborers, military
personnel, or athletes exercise strenuously in the heat. In addition to
the very young and very old, preadolescents and teenagers are at risk
since they may use poor judgment when vigorously exercising in high
humidity and heat. Other risk factors include obesity, poor conditioning with lack of acclimatization, and mild dehydration.
Cardiovascular inefficiency is a common feature of heat illness. Any
physiologic or pharmacologic impediment to cutaneous perfusion
impairs heat loss. Many patients are unaware of the heat risk associated with their medications. Anticholinergic agents impair sweating
and blunt the normal cardiovascular response to heat. Phenothiazines
and heterocyclic antidepressants also have anticholinergic properties
that interfere with the function of the preoptic nucleus of the anterior
hypothalamus due to central depletion of dopamine.
Calcium channel blockers, beta blockers, and various stimulants
also inhibit sweating by reducing peripheral blood flow. To maintain
the mean arterial blood pressure, increased cardiac output must be
capable of compensating for progressive dehydration. A variety of
stimulants and substances of abuse also increase muscle activity and
heat production.
Careful consideration of the differential diagnosis is important
in the evaluation of a patient for a potential heat-related illness. The
clinical setting may suggest other etiologies, such as malignant hyperthermia after general anesthesia. Neuroleptic malignant syndrome can
be triggered by certain antipsychotic medications, including selective
serotonin reuptake inhibitors. A variety of infectious and endocrine
disorders as well as toxicologic or CNS etiologies may mimic heatstroke (Table 465-1).
■ MINOR HEAT-EMERGENCY SYNDROMES
Heat edema is characterized by mild swelling of the hands, feet, and
ankles during the first few days of significant heat exposure. The principal mechanism involves cutaneous vasodilation and pooling of interstitial fluid in response to heat stress. Heat also increases the secretion
3636 PART 15 Disorders Associated with Environmental Exposures
or lotion provides some relief. In adults, localized areas may benefit
from 1% salicylic acid TID, with caution taken to avoid salicylate
intoxication. Clothing with breathable fabric should be clean and loose
fitting, and activities or environments that induce diaphoresis should
be avoided.
Heat syncope (exercise-associated collapse) can follow endurance
exercise or occur in the elderly. Other common clinical scenarios
include prolonged standing while stationary in the heat and sudden
standing after prolonged exposure to heat. Heat stress routinely causes
relative volume depletion, decreased vasomotor tone, and peripheral
vasodilation. The cumulative effect of this decrease in venous return
is postural hypotension, especially in nonacclimated elderly individuals. Many of those affected also have comorbidities. Therefore, other
cardiovascular, neurologic, and metabolic causes of syncope should
be considered. After removal from the heat source, most patients will
recover promptly with cooling and rehydration.
Hyperventilation tetany occurs in some individuals when exposure
to heat stimulates hyperventilation, producing respiratory alkalosis,
paresthesia, and carpopedal spasm. Unlike heat cramps, heat tetany
causes very little muscle-compartment pain. Treatment includes providing reassurance, moving the patient out of the heat, and addressing
the hyperventilation.
■ HEAT CRAMPS
Heat cramps (exercise-associated muscle cramps) are intermittent,
painful, and involuntary spasmodic contractions of skeletal muscles.
They typically occur in an unacclimated individual who is at rest after
vigorous exertion in a humid, hot environment. In contrast, cramps
that occur in athletes during exercise last longer, are relieved by stretching and massage, and resolve spontaneously.
Of note, not all muscle cramps are related to exercise, and the differential diagnosis includes many other disorders. A variety of medications, myopathies, endocrine disorders, and sickle cell trait are other
possible causes.
The typical patient with heat cramps is usually profusely diaphoretic
and has been replacing fluid losses with copious water or other hypotonic fluids. Roofers, firefighters, military personnel, athletes, steel
workers, and field workers are commonly affected. Other predisposing
factors include insufficient sodium intake before intense activity in the
heat and lack of heat acclimatization, resulting in sweat with a high salt
concentration.
The precise pathogenesis of heat cramps appears to involve a relative deficiency of sodium, potassium, and fluid at the intracellular
level. Coupled with copious hypotonic fluid ingestion, large amounts
of sodium in the diaphoresis cause hyponatremia and hypochloremia,
resulting in muscle cramps due to calcium-dependent muscle relaxation. Total-body depletion of potassium may be observed during the
period of heat acclimatization. Rhabdomyolysis is very rare with routine exercise-associated muscle cramps.
Heat cramps that are not accompanied by significant dehydration
can be treated with commercially available electrolyte solutions.
Although the flavored electrolyte solutions are far more palatable, two
650-mg salt tablets dissolved in 1 quart of water produce a 0.1% saline
solution. Individuals should avoid the ingestion of undissolved salt
tablets, which are a gastric irritant and may induce vomiting.
■ HEAT EXHAUSTION
The physiologic hallmarks of heat exhaustion—in contrast to
heatstroke—are the maintenance of thermoregulatory control and
CNS function. The core temperature is usually elevated but is generally
<40.5°C (<105°F). The two physiologic precipitants are water depletion
and sodium depletion, which often occur in combination. Laborers,
athletes, and elderly individuals exerting themselves in hot environments, without adequate fluid intake, tend to develop water-depletion
heat exhaustion. Persons working in the heat frequently consume only
two-thirds of their net water loss and are voluntarily dehydrated. In
contrast, salt-depletion heat exhaustion occurs more slowly in unacclimated persons who have been consuming large quantities of hypotonic
solutions.
TABLE 465-1 Heat-Related Illness: Predisposing Factors and
Differential Diagnosis
ILLNESS PREDISPOSING FACTORS
Cardiovascular inefficiency Age extremes
Beta/calcium channel blockade
Congestive heart failure
Dehydration
Diuresis
Obesity
Poor physical fitness
Central nervous system illness Cerebellar injury
Cerebral hemorrhage
Hypothalamic cerebrovascular accident
Psychiatric disorders
Status epilepticus
Impaired heat loss Antihistamines
Heterocyclic antidepressants
Occlusive clothing
Skin abnormalities
Endocrine and immune-related
illness
Diabetic ketoacidosis
Multiple-organ dysfunction syndrome
Pheochromocytoma
Systemic inflammatory response syndrome
Thyroid storm
Excessive heat load Environmental conditions
Exertion
Fever
Hypermetabolic state
Lack of acclimatization
Infectious illness Cerebral abscess
Encephalitis
Malaria
Meningitis
Sepsis syndrome
Tetanus
Typhoid
Toxicologic illness Amphetamines
Anticholinergic toxidrome
Cocaine
Dietary supplements
Hallucinogens
Malignant hyperthermia
Neuroleptic malignant syndrome
Salicylates
Serotonin syndrome
Strychnine
Sympathomimetics
Withdrawal syndromes (ethanol, hypnotics)
of antidiuretic hormone and aldosterone. Systemic causes of edema,
including cirrhosis, nephrotic syndrome, and congestive heart failure,
can usually be excluded by the history and physical examination. Heat
edema generally resolves without treatment in several days. Simple
leg elevation or compression stockings will usually suffice. Diuretics
are not effective and, in fact, predispose to volume depletion and the
development of more serious heat-related illnesses.
Prickly heat (miliaria rubra, lichen tropicus) is a maculopapular,
pruritic, erythematous rash that commonly occurs in clothed areas.
Blockage of the sweat pores by debris from macerated stratum corneum causes inflammation in the sweat ducts. As the ducts dilate, they
rupture and produce superficial vesicles. The predominant symptom is
pruritus. In addition to antihistamines, chlorhexidine in a light cream
3637Heat-Related Illnesses CHAPTER 465
Heat exhaustion is usually a diagnosis of exclusion because of the
multitude of nonspecific symptoms. If any signs of heatstroke are
present, rapid cooling and crystalloid resuscitation should be initiated
immediately during stabilization and evaluation. Mild neurologic
and gastrointestinal influenza-like symptoms are common. These
symptoms may include headache, vertigo, ataxia, impaired judgment,
malaise, dizziness, nausea, and muscle cramps. Orthostatic hypotension and sinus tachycardia develop frequently. More significant CNS
impairment suggests heatstroke or other infectious, neurologic, or
toxicologic diagnoses.
Hemoconcentration does not always develop, and rapid infusion of
isotonic IV fluids should be guided by frequent electrolyte determinations and perfusion requirements. Most cases of heat exhaustion reflect
mixed sodium and water depletion. Sodium-depletion heat exhaustion is characterized by hyponatremia and hypochloremia. Hepatic
aminotransferases are mildly elevated in both types of heat exhaustion.
Urinary sodium and chloride concentrations are usually low.
Some patients with heat exhaustion develop heatstroke after removal
from the heat-stress environment. Aggressive cooling of nonresponders is indicated until their core temperature is 39°C (102.2°F). Except
in mild cases, free water deficits should be replaced slowly over 24–48 h
to avoid a decrease of serum osmolality by >2 mOsm/h.
The disposition of younger, previously healthy heat-exhaustion
patients who have no major laboratory abnormalities may include
hospital observation and discharge after IV rehydration. Older patients
with comorbidities (including cardiovascular disease) or predisposing
factors often require inpatient fluid and electrolyte replacement, monitoring, and reassessment.
■ HEATSTROKE
The clinical manifestations of heatstroke reflect a total loss of thermoregulatory function. Typical vital-sign abnormalities include tachypnea, various tachycardias, hypotension, and a widened pulse
pressure. Although there is no single specific diagnostic test, the historical and physical triad of exposure to a heat stress, CNS dysfunction,
and a core temperature >40.5°C helps establish the preliminary diagnosis. Some patients with impending heatstroke will initially appear lucid.
The definitive diagnosis should be reserved until the other potential
causes of hyperthermia are excluded. Many of the usual laboratory
abnormalities seen with heatstroke overlap with other conditions. If
the patient’s mental status does not improve with cooling, toxicologic
screening may be indicated, and cranial CT and spinal fluid analysis
can be considered.
The premonitory clinical characteristics may be nonspecific and
include weakness, dizziness, disorientation, ataxia, and gastrointestinal
or psychiatric symptoms. These prodromal symptoms often resemble
heat exhaustion. The sudden onset of heatstroke occurs when the maintenance of adequate perfusion requires peripheral vasoconstriction to
stabilize the mean arterial blood pressure. As a result, the cutaneous
radiation of heat ceases. At this juncture, the core temperature rises
dramatically. Since many patients with heatstroke also meet the criteria
for systemic inflammatory response syndrome (SIRS) and have a broad
differential diagnosis, rapid cooling is essential during the extensive
diagnostic evaluation. Heat-induced SIRS reflects the responses of both
the innate and the adaptive immune systems (Table 465-1).
There are two forms of heatstroke with significantly different manifestations (Table 465-2). Classic (epidemic) heatstroke (CHS) usually
occurs during long periods of high ambient temperature and humidity,
as during summer heat waves. Patients with CHS commonly have
chronic diseases that predispose to heat-related illness, and they may
have limited access to oral fluids. Heat dissipation mechanisms are
overwhelmed by both endogenous heat production and exogenous heat
stress. Patients with CHS are often compliant with prescribed medications that can impair tolerance to a heat stress. In many of these dehydrated CHS patients, sweating has ceased and the skin is hot and dry.
If cooling is delayed, severe hepatic dysfunction, renal failure, disseminated intravascular coagulation, and fulminant multisystem organ
failure may occur. Hepatocytes are very heat sensitive. On presentation, the serum level of aspartate aminotransferase (AST) is routinely
elevated. Eventually, levels of both AST and alanine aminotransferase
(ALT) often increase to >100 times the normal values. Coagulation studies commonly demonstrate decreased platelets, fibrinogen,
and prothrombin. Most patients with CHS require cautious crystalloid resuscitation, electrolyte monitoring, and—in certain refractory
cases—consideration of central venous pressure (CVP) measurements.
Hypernatremia is secondary to dehydration in CHS. Many patients
exhibit significant stress leukocytosis, even in the absence of infection.
Patients with exertional heatstroke (EHS), in contrast to those with
CHS, are often young and previously healthy, and their diagnosis is
usually more obvious from the history. Athletes, laborers, and military recruits are common victims. Unlike those with CHS, many EHS
patients present profusely diaphoretic despite significant dehydration.
As a result of muscular exertion, rhabdomyolysis and acute renal failure are more common in EHS. Studies to detect rhabdomyolysis and
its complications, including hypocalcemia and hyperphosphatemia,
should be considered. Hyponatremia, hypoglycemia, and coagulopathies are frequent findings. Elevated creatine kinase and lactate dehydrogenase levels also suggest EHS. Oliguria is a common finding. Renal
failure can result from direct thermal injury, untreated rhabdomyolysis,
or volume depletion. Common urinalysis findings include microscopic
hematuria, myoglobinuria, and granular or red cell casts.
With both CHS and EHS, heat-related increases in cardiac biomarker
levels may be present and reversible. Heatstroke often causes thermal
cardiomyopathy. As a result, the CVP may be elevated despite significant dehydration. In addition, the patient often presents with potentially deceptive noncardiogenic pulmonary edema and basilar rales
despite being significantly hypovolemic. The electrocardiogram commonly displays a variety of tachyarrhythmias, nonspecific ST-T wave
changes, and heat-related ischemia or infarction. Rapid cooling—not
the initial administration of antiarrhythmic medications—is essential.
Above 42°C (107.6°F), heat can rapidly produce direct cellular
injury. Thermosensitive enzymes become nonfunctional, and eventually, there is irreversible uncoupling of oxidative phosphorylation. The
production of heat-shock proteins increases, and cytokines mediate
a systemic inflammatory response. The vascular endothelium is also
damaged, and this injury activates the coagulation cascade. Significant
shunting away from the splanchnic circulation produces gastrointestinal ischemia. Endotoxins further impair normal thermoregulation. As
a result, if cooling is delayed, severe hepatic dysfunction, permanent
renal failure, disseminated intravascular coagulation, and fulminant
multisystem organ failure may occur.
■ COOLING STRATEGIES
Before cooling is initiated, endotracheal intubation and continuous core-temperature monitoring should be considered. Peripheral
methods to measure temperature are not reliable. Hypoglycemia is
TABLE 465-2 Typical Manifestations of Heatstroke
CLASSIC EXERTIONAL
Older patient Younger patient
Predisposing health factors/
medications
Healthy condition
Epidemiology (heat waves) Sporadic cases
Sedentary Exercising
Anhidrosis (possible) Diaphoresis (common)
Central nervous system dysfunction Myocardial/hepatic injury
Oliguria Acute renal failure
Coagulopathy (mild) Disseminated intravascular
coagulation
Mild lactic acidosis Marked lactic acidosis
Mild creatine kinase elevation Rhabdomyolysis
Normoglycemia/calcemia Hypoglycemia/calcemia
Normokalemia Hyperkalemia
Normonatremia Hyponatremia
3638 PART 15 Disorders Associated with Environmental Exposures
a frequent finding and can be addressed by glucose infusion. Since
peripheral vasoconstriction delays heat dissipation, repeated administration of discrete boluses of isotonic crystalloid for hypotension is
preferable to the administration of α-adrenergic agonists.
Evaporative cooling is frequently the most practical and effective
technique. Rapid cooling is essential in both CHS and EHS, and an
immediate improvement in vital signs and mental status may prove
valuable for diagnostic purposes. Cool water (15°C [60°F]) is sprayed
on the exposed skin while fans direct continuous airflow over the
moistened skin. Cold packs applied to the neck, axillae, and groin are
useful cooling adjuncts. If cardiac electrodes will not adhere, they can
be applied to the patient’s back.
Immersion cooling in ice-cold water is an alternative option in EHS
but can induce peripheral vasoconstriction and shivering. The initial
increase in temperature from peripheral vasoconstriction will rapidly
be overcome by the large conductive thermal transfer into cold water.
This technique presents significant monitoring and resuscitation challenges in many clinical settings. The safety of immersion cooling is
best established for young, previously healthy patients with EHS (but
not for those with CHS). To avoid hypothermic afterdrop (continued
cooling after immersion), active cooling should be terminated at
~38°–39°C (100.4°F–102.2°F).
Cooling with commercially available cooling blankets should not be
the sole technique used, since the rate of cooling is far too slow. Other
methods are less efficacious and rarely indicated, such as IV infusion
of cold fluids and cold irrigation of the bladder or gastrointestinal
tract. Cold thoracic and peritoneal lavage are efficient maneuvers but
are invasive and rarely necessary. Endovascular cooling also provides
effective cooling.
■ RESUSCITATION
Aspiration commonly occurs in heatstroke, and endotracheal intubation is usually necessary. Depolarizing agents should be avoided. The
metabolic demands are high, and supplemental oxygenation is essential
due to hypoxemia induced by thermal stress and pulmonary dysfunction. The oxyhemoglobin dissociation curve is shifted to the right.
Pneumonitis, pulmonary infarction, hemorrhage, edema, and acute
respiratory distress syndrome occur frequently in heatstroke patients.
Seizures are common and can occur during therapeutic cooling. Cold
induced tonic-clonic muscular rigidity mimics seizure activity.
The circulatory fluid requirements, particularly in CHS, may be
deceptively modest. Aggressive cooling and modest volume repletion
usually elevate the CVP to 12–14 mmHg. The reading, however, may
be deceptive. Many patients present with a thermally induced hyperdynamic circulation accompanied by a high cardiac index, low peripheral
vascular resistance, and an elevated CVP caused by right-sided heart
failure. In contrast, most patients with EHS require far more zealous
isotonic crystalloid resuscitation.
The hypotension that is initially common among patients with heatstroke results from both dehydration and high-output cardiac failure
caused by peripheral vasodilation. Inotropes causing α-adrenergic
stimulation (e.g., norepinephrine) can impede cooling by causing significant vasoconstriction. Vasoactive catecholamines such as dopamine or dobutamine may be necessary if the cardiac output remains
depressed despite an elevated CVP, particularly in patients with a
hyperdynamic circulation.
A wide variety of tachyarrhythmias are routinely observed on
presentation and usually resolve spontaneously during cooling. The
administration of atrial or ventricular antiarrhythmic medications is
rarely indicated during cooling. Anticholinergic medications (including atropine) inhibit sweating and should be avoided. With a cardiac
rhythm that sustains perfusion, electrical cardioversion of the hyperthermic myocardium should be deferred until the myocardium is
cooled. Significant shivering, discomfort, or extreme agitation is preferably mitigated with short-acting benzodiazepines, which are ideal
due to their renal clearance. On the other hand, chlorpromazine may
lower the seizure threshold, has anticholinergic properties, and can
exacerbate the hypotension or cause neuroleptic malignant syndrome.
With hepatic dysfunction, barbiturates should be avoided and seizures
treated with benzodiazepines.
Coagulopathies more commonly occur after the first day of illness. After cooling, the patient should be monitored for disseminated
intravascular coagulation, and replacement therapy with fresh-frozen
plasma and platelets should be considered.
There is no therapeutic role for antipyretics in the control of environmentally induced hyperthermia; these drugs block the actions of
pyrogens at hypothalamic receptor sites. Salicylates can further uncouple oxidative phosphorylation in heatstroke and exacerbate coagulopathies. Acetaminophen may further stress hepatic function. The safety
and efficacy of dantrolene are not established. Although aminocaproic
acid impedes fibrinolysis, it may cause rhabdomyolysis and is not recommended in heatstroke.
■ DISPOSITION
Most patients with minor heat-emergency syndromes (including heat
edema, heat syncope, and heat cramps) require only stabilization and
treatment with outpatient follow-up. Although there are no decision
rules to guide disposition choices in heat exhaustion, many of these
patients have multiple predisposing factors and comorbidities that will
require prolonged observation or hospital admission.
Essentially all patients with actual heatstroke require admission to a
monitored setting, and most require intensive care. There are reports
of very high survival rates of patients following prehospital immersion
cooling without intensive care. Most or all of these patients appear to
have had heat exhaustion. Many actual heatstroke patients also require
prolonged tracheal intubation, invasive hemodynamic monitoring,
and support for various degrees of multiorgan dysfunction syndrome.
The prognosis worsens if the initial core temperature exceeds 42°C
(107.6°F) or if there was a prolonged period during which the core
temperature exceeded this level. Other features of a negative prognosis
include acute renal failure, massively elevated liver enzymes, and significant hyperkalemia. As expected, the number of dysfunctional organ
systems also correlates directly with mortality risk.
■ FURTHER READING
Balmain BN et al: Aging and thermoregulatory control: The clinical
implications of exercising under heat stress in older individuals.
BioMed Res Int 2018:8306154, 2018.
Casa DJ et al: National Athletic Trainers’ Association position statement: Exertional heat illnesses. J Athl Train 50:986, 2015.
Hosokawa Y et al: Inconsistency in the standard of care-toward
evidence-based management of exertional heat stroke. Front Physiol
10:108, 2019.
Lawton EM et al: Review article: Environmental heatstroke and
long-term clinical neurological outcomes: A literature review of case
reports and case series 2000-2016. Emerg Med Australas 31:163,
2019.
Leon LR et al: Pathophysiology of heat-related illnesses, in Auerbach’s
Wilderness Medicine, 7th ed. PS Auerbach et al (eds): Philadelphia,
Elsevier, 2017, pp. 249–267.
Lipman GS et al: Wilderness Medical Society practice guidelines for
the prevention and treatment of heat-related illness. Wilderness
Environ Med 24:351, 2013.
Platt M et al: Heat illness, in Rosen’s Emergency Medicine: Concepts
and Clinical Practice, 9th ed. Walls RM et al (eds). Philadelphia,
Elsevier, 2018, pp. 1755–1764.
Genes, the Environment, and Disease PART 16
Principles of Human
Genetics
J. Larry Jameson, Peter Kopp
466
IMPACT OF GENETICS AND GENOMICS
ON MEDICAL PRACTICE
Human genetics refers to the study of individual genes, their role
and function in disease, and their mode of inheritance. Genomics
refers to an organism’s entire genetic information, the genome, and
the function and interaction of DNA within the genome, as well as
with environmental or nongenetic factors, such as a person’s lifestyle.
With the characterization of the human genome, genomics not only
complements traditional genetics in our efforts to elucidate the etiology and pathogenesis of disease, but it now plays a prominent and
continuously expanding role in diagnostics, prevention, and therapy
(Chap. 467). These transformative developments, originally emerging from the Human Genome Project, have been variably designated
genomic medicine, personalized medicine, or precision medicine. Precision medicine aims at customizing medical decisions to an individual
patient. For example, a patient’s genetic characteristics (genotype)
can be used to optimize drug therapy and predict efficacy, adverse
events, and drug dosing of selected medications (pharmacogenomics)
(Chap. 68). The characterization of the mutational profile of a malignancy allows the identification of driver mutations or overexpressed
signaling molecules, which then facilitates the selection of targeted
therapies. Genome-wide polygenic risk scores (PRS) for common
complex diseases are also beginning to emerge and potentially impact
disease prevention.
Genetics has traditionally been viewed through the window of
relatively rare single-gene diseases. These disorders account for
~10% of pediatric admissions and childhood mortality. Historically,
genetics has focused predominantly on chromosomal and metabolic
disorders, reflecting the long-standing availability of techniques to
diagnose these conditions. For example, conditions such as trisomy
21 (Down’s syndrome) or monosomy X (Turner’s syndrome) can be
diagnosed using cytogenetics. Likewise, many metabolic disorders
(e.g., phenylketonuria, familial hypercholesterolemia) are diagnosed
using biochemical analyses. The advances in DNA and RNA diagnostics have extended the field of genetics to include virtually all medical
specialties and have led to the elucidation of the pathogenesis of the
majority of monogenic disorders. In addition, it is apparent that virtually every medical condition has a genetic component. As is often
evident from a patient’s family history, many common disorders such
as hypertension, heart disease, asthma, diabetes mellitus, and mental
illnesses are significantly influenced by the genetic background. These
polygenic or multifactorial (complex) disorders involve the contributions of many different genes, as well as environmental factors that can
modify disease risk. Genome-wide association studies (GWAS) have
elucidated numerous disease-associated loci and are providing novel
insights into the allelic architecture of complex traits. These studies
have been facilitated by the availability of comprehensive catalogues
of human single nucleotide polymorphism (SNP) haplotypes (HapMap, International Genome Sample Resource/1000 Genomes Project).
Next-generation DNA sequencing (NGS) technologies have evolved
rapidly, and the cost of sequencing whole exomes (the exons within
the genome; whole exome sequencing [WES]) or genomes (whole
genome sequencing [WGS]) has plummeted. Comprehensive unbiased
sequence analyses are now frequently used to characterize individuals
with complex undiagnosed conditions or to determine the mutational
profile of advanced malignancies in order to select targeted therapies.
The routine assembly of diploid genomes, which can reveal a comprehensive spectrum of human genetic variation, will be possible in the
near future and should provide further insights into heritability and
disease mechanisms.
Cancer has a genetic basis because it results from acquired somatic
mutations in genes controlling growth, apoptosis, and cellular differentiation (Chap. 71). In addition, the development of many cancers
is associated with a hereditary predisposition. Characterization of the
genome (and epigenome) in various malignancies has led to fundamental new insights into cancer biology and reveals that the genomic
profile of mutations is in many cases more important in determining
the appropriate therapy than the organ in which the tumor originates.
The Cancer Genome Atlas (TCGA) initiative of the National Cancer
Institute and the National Human Genome Research Institute has
already characterized the genomic landscape of >30 malignancies.
TCGA consists of comprehensive analyses of genomic and proteomic
alterations and is providing fundamental new insights into the molecular pathogenesis of cancer. These data, together with comprehensive
catalogues of somatic mutations identified in human cancer, have
direct clinical ramifications that impact cancer taxonomy, as well as the
development and choice of targeted therapies.
Genetic and genomic approaches have proven invaluable for the
detection of infectious pathogens and are used clinically to identify
agents that are difficult to culture such as mycobacteria, viruses, and
parasites, or to track infectious agents locally or globally. In many
cases, molecular genetics has improved the feasibility and accuracy of
diagnostic testing and is beginning to open new avenues for therapy,
including gene and cellular therapies (Chap. 470). Molecular genetics
has also provided the opportunity to characterize the microbiome,
a new field that characterizes the population dynamics of bacteria,
viruses, and parasites that coexist with humans and other animals
(Chap. 471). Emerging data indicate that the microbiome has significant effects on normal physiology as well as various disease states, and
the field is now focusing on defining the mechanisms underlying these
interactions.
Molecular biology has significantly changed the treatment of human
disease. Peptide hormones, growth factors, cytokines, and vaccines
can be produced in large amounts using recombinant DNA and RNA
technology (e.g., mRNA vaccines against SARS-CoV-2). Targeted
modifications of recombinant peptides provide improved therapeutic
tools, as illustrated by genetically modified insulin analogues with
more favorable kinetics.
The astounding rate at which new genetic and genomic information
is being generated has led to major challenges for physicians, health
care providers, and basic investigators. Although many functional
aspects of the genome remain unknown, there are many clinical
situations where sufficient evidence exists for the use of genetic and
genomic information to optimize patient care and treatment. Much
genetic information resides in databases that provide easy access to
the expanding information about the human genome, genetic disease,
and genetic testing (Table 466-1). For example, several thousand
monogenic disorders are summarized in a large, continuously evolving
compendium, referred to as the Online Mendelian Inheritance in Man
(OMIM) catalogue (Table 466-1). The constant refinement of bioinformatics and new developments in big data analytics, together with the
widespread adoption of electronic health records (EHRs), are simplifying the access, analysis, and integration of this daunting amount of new
information. Importantly, genomic data can be integrated readily into
EHRs and thus impact clinical practice.
■ THE HUMAN GENOME
Structure of the Human Genome The Human Genome Project
was initiated in the mid-1980s as an ambitious effort to characterize the
entire human genome and culminated in the completion of the DNA
3640 PART 16 Genes, the Environment, and Disease
TABLE 466-1 Selected Databases Relevant for Genomics and Genetic Disorders
SITE URL COMMENT
National Center for Biotechnology
Information (NCBI)
http://www.ncbi.nlm.nih.gov/ Broad access to biomedical and genomic information, literature (PubMed),
sequence databases, software for analyses of nucleotides and proteins
Extensive links to other databases, genome resources, and tutorials
National Human Genome Research
Institute
http://www.genome.gov/ An institute of the National Institutes of Health focused on genomic and genetic
research; links providing information about the human genome sequence,
genomes of other organisms, and genomic research
Catalog of Published Genome-Wide
Association Studies
https://www.ebi.ac.uk/gwas/ Published high-resolution genome-wide association studies (GWAS)
Ensembl Genome browser http://www.ensembl.org Maps and sequence information of eukaryotic genomes
Online Mendelian Inheritance in Man http://www.ncbi.nlm.nih.gov/omim Online compendium of Mendelian disorders and human genes causing genetic
disorders
American College of Medical Genetics and
Genomics
http://www.acmg.net/ Extensive links to other databases relevant for the diagnosis, treatment, and
prevention of genetic disease
American Society of Human Genetics http://www.ashg.org Information about advances in genetic research, professional and public
education, and social and scientific policies
The Cancer Genome Atlas https://cancergenome.nih.gov/ Comprehensive, multidimensional characterization of the genomic and proteomic
landscape of malignancies with high public health impact
COSMIC Catalogue of Somatic Mutations
in Cancer
https://cancer.sanger.ac.uk/cosmic Comprehensive catalogue of somatic mutations in human cancer
Genetic Testing Registry https://www.ncbi.nlm.nih.gov/gtr/ International directory of genetic testing laboratories and prenatal diagnosis
clinics; reviews and educational materials
Genomes Online Database (GOLD) http://www.genomesonline.org/ Information on published and unpublished genomes
HUGO Gene Nomenclature http://www.genenames.org/ Gene names and symbols
GENECODE https://www.gencodegenes.org/ High-quality reference gene annotation and experimental validation for human and
mouse genomes
MITOMAP, a human mitochondrial genome
database
http://www.mitomap.org/ A compendium of polymorphisms and mutations of the human mitochondrial DNA
The International Genome Sample
Resource (IGSR)
http://www.internationalgenome.org Public catalogue of human variation and genotype data from numerous ethnic
groups
Human Genome Variation Society https://www.hgvs.org/ Collection and documentation of genomic variations including population
distribution and phenotypic associations
ENCODE http://www.genome.gov/10005107 Encyclopedia of DNA Elements; catalogue of all functional elements in the human
genome
Dolan DNA Learning Center, Cold Spring
Harbor Laboratories
http://www.dnalc.org/ Educational material about selected genetic disorders, DNA, eugenics, and
genetic origin
The Online Metabolic and Molecular
Bases of Inherited Disease (OMMBID)
http://ommbid.mhmedical.com Online version of the comprehensive text on the metabolic and molecular bases of
inherited disease
Online Mendelian Inheritance in Animals
(OMIA)
https://www.omia.org/home/ Online compendium of Mendelian disorders in animals
The Jackson Laboratory http://www.jax.org/ Information about murine models and the mouse genome
Mouse genome informatics http://www.informatics.jax.org Mouse genome informatics, potential mouse models of human disease, information
on phenotypic similarity between mouse models and human patients
Note: Databases are evolving constantly. Pertinent information may be found by using links listed in the few selected databases.
sequence for the last of the human chromosomes in 2006. The scope of
a whole genome sequence analysis can be illustrated by the following
analogy. Human DNA consists of ~3 billion base pairs (bp) of DNA
per haploid genome, which is nearly 1000-fold greater than that of the
Escherichia coli genome. If the human DNA sequence were printed
out, it would correspond to about 120 volumes of Harrison’s Principles
of Internal Medicine.
In addition to the human genome, the genomes of thousands of
organisms have been sequenced completely or partially (Genomes
Online Database [GOLD]; Table 466-1). They include, among others,
eukaryotes such as the mouse (Mus musculus), Saccharomyces cerevisiae, Caenorhabditis elegans, and Drosophila melanogaster; bacteria
(e.g., E. coli); and archaea, viruses, organelles (mitochondria, chloroplasts), and plants (e.g., Arabidopsis thaliana). Genomic information
of infectious agents has significant impact for the characterization
of infectious outbreaks and epidemics. Other ramifications arising
from the availability of genomic data include, among others, (1) the
comparison of entire genomes (comparative genomics); (2) the study
of large-scale expression of RNAs (functional genomics), proteins
(proteomics), or protein families (e.g., the kinome, the complete set of
protein kinases) to detect differences between various tissues in health
and disease; (3) the characterization of the variation among individuals by establishing catalogues of sequence variations and SNPs; and
(4) the identification of genes that play critical roles in the development
of polygenic and multifactorial disorders.
CHROMOSOMES The human genome is divided into 23 different chromosomes, including 22 autosomes (numbered 1–22) and the X and Y sex
chromosomes (Fig. 466-1). Adult cells are diploid, meaning they contain
two homologous sets of 22 autosomes and a pair of sex chromosomes.
Females have two X chromosomes (XX), whereas males have one X
and one Y chromosome (XY). As a consequence of meiosis, germ cells
(sperm or oocytes) are haploid and contain one set of 22 autosomes and
one of the sex chromosomes. At the time of fertilization, the diploid
genome is reconstituted by pairing of the homologous chromosomes
from the mother and father. With each cell division (mitosis), chromosomes are replicated, paired, segregated, and divided into two daughter
cells.
Principles of Human Genetics
3641CHAPTER 466
STRUCTURE OF DNA DNA is a double-stranded helix composed of
four different bases: adenine (A), thymidine (T), guanine (G), and
cytosine (C). Adenine is paired to thymidine, and guanine is paired
to cytosine, by hydrogen bond interactions that span the double helix
(Fig. 466-1). DNA has several remarkable features that make it ideal
for the transmission of genetic information. It is relatively stable, and
the double-stranded nature of DNA and its feature of strict base-pair
complementarity permit faithful replication during cell division. Complementarity also allows the transmission of genetic information from
DNA → RNA → protein (Fig. 466-2). mRNA is encoded by the socalled sense or coding strand of the DNA double helix and is translated
into proteins by ribosomes.
The presence of four different bases provides surprising genetic
diversity. In the protein-coding regions of genes, the DNA bases are
arranged into codons, a triplet of bases that specifies a particular
amino acid. It is possible to arrange the four bases into 64 different
triplet codons (43
). Each codon specifies 1 of the 20 different amino
acids, or a regulatory signal such as initiation and stop of translation.
Because there are more codons than amino acids, the genetic code is
degenerate; that is, most amino acids can be specified by several different codons. By arranging the codons in different combinations and
in various lengths, it is possible to generate the tremendous diversity of
primary protein structure.
DNA length is normally measured in units of 1000 bp (kilobases, kb)
or 1,000,000 bp (megabases, Mb). In the human genome, only ~1% of
DNA accounts for protein-coding sequences. The noncoding DNA has
multiple functional and structural roles including (1) sequences that
form introns; (2) regulatory elements (promoters, enhancers, silencers,
insulators); (3) sequences that generate RNAs that do not code for proteins; (4) centromeres and telomeres; (5) regions defining chromatin
structure and histone modifications; (6) various forms of repetitive
sequences of variable length; and (7) pseudogenes and regions without
currently discernible functional or structural roles (Fig. 466-1).
GENES A gene is a functional unit that is regulated by transcription
(see below) and encodes an RNA product, which is most commonly,
but not always, translated into a protein that exerts activity within
or outside the cell (Fig. 466-3). Historically, genes were identified
because they conferred specific traits that are transmitted from one
generation to the next. Now, they are frequently characterized based
on expression in various tissues (transcriptome). The size of genes is
quite broad; some genes are only a few hundred base pairs, whereas
others are extraordinarily large (2 Mb). The number of genes greatly
underestimates the complexity of genetic expression, because single
genes can generate multiple spliced messenger RNA (mRNA) products
(isoforms), which are translated into proteins that are subject to complex posttranslational modification such as phosphorylation. Exons
refer to the portion of genes that are eventually spliced together to form
mRNA. Introns refer to the spacing regions between the exons that
are spliced out of precursor RNAs during RNA processing. The gene
locus also includes regions that are necessary to control its expression
(Fig. 466-2). Current estimates predict roughly 20,000 protein-coding
genes in the human genome with an average of about four different
coding transcripts per gene. Remarkably, the exome only constitutes
1.14% of the genome. Of note, the number of transcripts is close to
200,000 and includes thousands of noncoding transcripts (RNAs of
various length such as microRNAs [miRNA] and long noncoding
RNAs [lncRNA]). These noncoding RNAs are involved in numerous
cellular processes such as transcriptional and posttranscriptional
regulation of gene expression, chromatin remodeling, and protein
trafficking, among others. Not surprisingly, aberrant expression and/or
mutations in these RNAs play a pathogenic role in numerous diseases.
SINGLE-NUCLEOTIDE POLYMORPHISMS Each individual has roughly
5 million sequence variants that differentiate one person from another.
Some of these variants have no impact on health, whereas others may
increase or lower the risk for developing a specific disease. Remarkably,
however, the primary DNA sequence of humans has ~99.9% similarity
compared to that of any other human. An SNP is a variation of a single base pair in the DNA. The identification of the ~10 million SNPs
estimated to occur in the human genome has generated a catalogue of
common genetic variants that occur in human beings from distinct
ethnic backgrounds (Fig. 466-3). SNPs are the most common type of
sequence variation and account for ~90% of all sequence variation.
They occur on average every 100–300 bases and are the major source
of genetic heterogeneity. SNPs that are in close proximity are inherited together (e.g., they are linked) and are referred to as haplotypes
(Fig. 466-4). Haplotype maps describe the nature and location of these
SNP haplotypes and how they are distributed among individuals within
and among populations, information that has been facilitating GWAS
designed to elucidate the complex interactions among multiple genes
and lifestyle factors in multifactorial disorders (see below). Moreover,
haplotype analyses are useful to assess variations in responses to medications (pharmacogenomics) and environmental factors, as well as the
prediction of disease predisposition.
COPY NUMBER VARIATIONS Copy number variations (CNVs) are relatively large genomic regions (1 kb to several Mb) that have been duplicated or deleted on certain chromosomes and hence alter the diploid
status of the DNA (Fig. 466-5). It has been estimated that 5–10% of the
genome can display CNVs. When comparing the genomes of two individuals, ~0.4–0.8% of their genomes differ in terms of CNVs scattered
throughout the genome. Of note, de novo CNVs have been observed
between monozygotic twins, who otherwise have identical genomes.
Some CNVs have no functional consequences, whereas others have
been associated with susceptibility or resistance to disease, and CNVs
also occur in cancer cells.
Histone
H1
Nucleosome
fiber
Metaphase
chromosome
Solenoid
q, long arm
p, short arm
Centromere
Supercoiled
chromatin
Telomere
Nucleosome core
Histone H2A, H2B, H4
Double-strand DNA
without histones
Adenine
H
H
H
O
O
P OO
O
O–
P OO
O
O–
O
H
H
H
H H
H
H
H
H
H H3C
H
Thymine
Cytosine Guanine
C C
C C
C
C
C
C
C C
C
C
C C C
C
N
N
T
A
C
G
G
C
A
T
N
N
N N
N
N N
N
N
C
C
C
N
N
N
N
FIGURE 466-1 Structure of chromatin and chromosomes. Chromatin is composed of
double-strand DNA that is wrapped around histone and nonhistone proteins forming
nucleosomes. The nucleosomes are further organized into solenoid structures.
Chromosomes assume their characteristic structure, with short (p) and long (q)
arms at the metaphase stage of the cell cycle.
3642 PART 16 Genes, the Environment, and Disease
CRE
Enhancer Silencer
RE CAAT TATA 1 2 3
1 2 3
Nuclear
receptor
Transcription
factor
RNA polymerase II
Nuclear
receptor
CBP TAF
GTF CREB CREB TBP
HAT
CoA
Steroids Ca Growth 2+ Cytokines
factors
Hormones
Light
UV-light
Mechanical stress
Regulation of Gene Expression
Transcription
mRNA Processing
Translation
Posttranslational Processing
Cytoplasm
Nucleus
DNA
hRNA
–COOH
5′ -Cap –Poly-A Tail
mRNA
Protein
1 2 3
NH2–
FIGURE 466-2 Flow of genetic information. Multiple extracellular signals activate intracellular signal cascades that result in altered regulation of gene expression through
the interaction of transcription factors with regulatory regions of genes. RNA polymerase transcribes DNA into RNA that is processed to mRNA by excision of intronic
sequences. The mRNA is translated into a polypeptide chain to form the mature protein after undergoing posttranslational processing. CBP, CREB-binding protein; CoA,
co-activator; COOH, carboxyterminus; CRE, cyclic AMP responsive element; CREB, cyclic AMP response element–binding protein; GTF, general transcription factors; HAT,
histone acetyl transferase; NH2, aminoterminus; RE, response element; TAF, TBP-associated factors; TATA, TATA box; TBP, TATA-binding protein.
Replication of DNA and Mitosis Genetic information in DNA
is transmitted to daughter cells under two different circumstances:
(1) somatic cells divide by mitosis, allowing the diploid (2n) genome
to replicate itself completely in conjunction with cell division; and (2)
germ cells (sperm and ova) undergo meiosis, a process that enables
the reduction of the diploid (2n) set of chromosomes to the haploid
state (1n).
Prior to mitosis, cells exit the resting, or G0
state, and enter the cell
cycle. After traversing a critical checkpoint in G1
, cells undergo DNA
synthesis (S phase), during which the DNA in each chromosome is replicated, yielding two pairs of sister chromatids (2n → 4n). The process
of DNA synthesis requires stringent fidelity in order to avoid transmitting errors to subsequent generations of cells. Genetic abnormalities
of DNA mismatch/repair include xeroderma pigmentosum, Bloom’s
syndrome, ataxia telangiectasia, and hereditary nonpolyposis colon
cancer (HNPCC), among others. Many of these disorders strongly
predispose to neoplasia because of the rapid acquisition of additional
mutations (Chap. 71). After completion of DNA synthesis, cells enter
G2
and progress through a second checkpoint before entering mitosis.
At this stage, the chromosomes condense and are aligned along the
equatorial plate at metaphase. The two identical sister chromatids, held
together at the centromere, divide and migrate to opposite poles of the
cell. After formation of a nuclear membrane around the two separated
sets of chromatids, the cell divides and two daughter cells are formed,
thus restoring the diploid (2n) state.
Assortment and Segregation of Genes During Meiosis Meiosis occurs only in germ cells of the gonads. It shares certain features
with mitosis but involves two distinct steps of cell division that reduce
the chromosome number to the haploid state. In addition, there is
active recombination that generates genetic diversity. During the first
cell division, two sister chromatids (2n → 4n) are formed for each chromosome pair and there is an exchange of DNA between homologous
paternal and maternal chromosomes. This process involves the formation of chiasmata, structures that correspond to the DNA segments that
cross over between the maternal and paternal homologues (Fig. 466-6).
Usually there is at least one crossover on each chromosomal arm;
recombination occurs more frequently in female meiosis than in male
meiosis. Subsequently, the chromosomes segregate randomly. Because
there are 23 chromosomes, there exist 223 (>8 million) possible combinations of chromosomes. Together with the genetic exchanges that
occur during recombination, chromosomal segregation generates tremendous diversity, and each gamete is genetically unique. The process
of recombination and the independent segregation of chromosomes
provide the foundation for performing linkage analyses, whereby one
attempts to correlate the inheritance of certain chromosomal regions
(or linked genes) with the presence of a disease or genetic trait (see
below).
After the first meiotic division, which results in two daughter cells
(2n), the two chromatids of each chromosome separate during a second meiotic division to yield four gametes with a haploid state (1n).
When the egg is fertilized by sperm, the two haploid sets are combined,
thereby restoring the diploid state (2n) in the zygote.
■ REGULATION OF GENE EXPRESSION
Regulation by Transcription Factors The expression of genes
is regulated by DNA-binding proteins that activate or repress transcription. The number of DNA sequences and transcription factors
that regulate transcription is much greater than originally anticipated.
Most genes contain at least 15–20 discrete regulatory elements within
300 bp of the transcription start site. This densely packed promoter
region often contains binding sites for ubiquitous transcription factors. However, factors involved in cell-specific expression may also
bind to these sequences. Key regulatory elements may also reside
at a large distance from the proximal promoter. The globin and the
Principles of Human Genetics
3643CHAPTER 466
Chromosome 7
p22.3
p22.1
p21.3
p21.1
p15.3
p15.1
p14.3
p14.1
p13
p12.3
p12.1
p1
q11.21
q11.22
q11.23
q21.11
p21.13
q21.3
q22.1
q22.3
q31.1
q31.2
q31.31
q31.33
q32.1
q33
q34
q35
q36.1
q36.3
1.2
Known Genes
(1260)
SNPs
(612,977)
CFTR Gene
116.90 Mb 116.94 Mb 116.98 Mb 117.02 Mb 117.06 Mb
200 Kb
20 Kb
SNPs
Intronic
Coding region, synonymous Coding region, nonsynonymous
Splice site
Coding region, frameshift
FIGURE 466-3 Chromosome 7 is shown with the density of single nucleotide polymorphisms (SNPs) and genes above. A 200-kb region in 7q31.2 containing the CFTR gene
is shown below. The CFTR gene contains 27 exons. Close to 2000 mutations in this gene have been found in patients with cystic fibrosis. A 20-kb region encompassing exons
4–9 is shown further amplified to illustrate the SNPs in this region.
FIGURE 466-4 The origin of haplotypes is due to repeated recombination events
occurring in multiple generations. Over time, this leads to distinct haplotypes. These
haplotype blocks can often be characterized by genotyping selected Tag single
nucleotide polymorphisms (SNPs), an approach that facilitates performing genomewide association studies (GWAS).
immunoglobulin genes, for example, contain locus control regions that
are several kilobases away from the structural sequences of the gene.
Specific groups of transcription factors that bind to these promoter
and enhancer sequences provide a combinatorial code for regulating
transcription. In this manner, relatively ubiquitous factors interact
with more restricted factors to allow each gene to be expressed and
regulated in a unique manner that is dependent on developmental
state, cell type, and numerous extracellular stimuli. Regulatory factors
also bind within the gene itself, particularly in the intronic regions.
The transcription factors that bind to DNA actually represent only the
first level of regulatory control. Other proteins—co-activators and corepressors—interact with the DNA-binding transcription factors to generate large regulatory complexes. These complexes are subject to control
by numerous cell-signaling pathways and enzymes, leading to phosphorylation, acetylation, sumoylation, and ubiquitination. Ultimately,
the recruited transcription factors interact with, and stabilize, components of the basal transcription complex that assembles at the site of the
TATA box and initiator region. This basal transcription factor complex
consists of >30 different proteins. Gene transcription occurs when
RNA polymerase begins to synthesize RNA from the DNA template.
A large number of identified genetic diseases involve transcription
factors (Table 466-2).
The field of functional genomics is based on the concept that understanding alterations of gene expression under various physiologic and
pathologic conditions provides insight into the underlying functional
3644 PART 16 Genes, the Environment, and Disease
1
2
Normal
Deleted
Area
Duplicated
Area
0
–1
–2
log2 (ratio)
Chromosome 8
FIGURE 466-5 Copy number variations (CNV) encompass relatively large regions of the genome that
have been duplicated or deleted. Chromosome 8 is shown with a CNV detected by genomic hybridization.
An increase in the signal strength indicates a duplication, whereas a decrease reflects a deletion of the
covered chromosomal regions.
Homologous
chromosomes
A
B
C
D
a
b
c
d
a
b
c
d
a
b
c
d
Chromatids
A
B
C
D
A
B
C
D
a
b
c
d
No crossover
A
B
C
D
a
b
c
d
A
B
C
D
a
b
C
d
Double crossover
A
B
C
D
a
b
c
d
A
B
c
D
a
b
C
D
Crossover
A
B
C
D
a
b
c
d
A
B
c
d
a
b
c
d
No recombination
in gametes
A
B
C
D
a
b
c
d
A
B
C
D
a
b
C
D
Recombination
in gametes
A
B
C
D
a
b
c
d
A
B
c
d
a
b
C
d
Recombination
in gametes
A
B
C
D
a
b
c
d
A
B
c
D
FIGURE 466-6 Crossing-over and genetic recombination. During chiasma
formation, either of the two sister chromatids on one chromosome pairs with
one of the chromatids of the homologous chromosome. Genetic recombination
occurs through crossing-over and results in recombinant and nonrecombinant
chromosome segments in the gametes. Together with the random segregation of
the maternal and paternal chromosomes, recombination contributes to genetic
diversity and forms the basis of the concept of linkage.
role of the gene. The ENCODE (Encyclopedia of DNA Elements) project aims to compile and annotate all functional sequences in the human
genome. By revealing specific gene expression profiles, this knowledge
can be of diagnostic and therapeutic relevance. The large-scale study
of expression profiles is referred to as transcriptomics because the
complement of mRNAs transcribed by the cellular genome is called
the transcriptome.
Most studies of gene expression have focused on the regulatory
DNA elements of genes that control transcription. However, it should
be emphasized that gene expression requires a series of steps, including
mRNA processing, protein translation, and posttranslational modifications, all of which are actively regulated (Fig. 466-2).
Epigenetic Regulation of Gene Expression (see Chap. 483)
Epigenetics describes mechanisms and phenotypic changes that are not
a result of variation in the primary DNA nucleotide sequence but are
caused by secondary modifications of DNA or histones. These modifications include heritable changes such as X-inactivation and imprinting, but they can also result from dynamic posttranslational protein
modifications in response to environmental influences such as diet,
age, or drugs. The epigenetic modifications result in altered expression
of individual genes or chromosomal loci encompassing multiple genes.
The term epigenome describes the constellation of covalent modifications of DNA and histones that impact chromatin structure, as well
as noncoding transcripts that modulate the transcriptional activity of
DNA. Although the primary DNA sequence is usually identical in all
cells of an organism, tissue-specific changes in the epigenome contribute to determining the transcriptional signature of a cell (transcriptome) and hence the protein expression profile (proteome).
Mechanistically, DNA and histone modifications can result in the
activation or silencing of gene expression (Fig. 466-7). DNA methylation involves the addition of a methyl group to cytosine residues.
This is usually restricted to cytosines of CpG dinucleotides, which
are abundant throughout the genome. Methylation of these dinucleotides is thought to represent a defense mechanism that minimizes the
expression of sequences that have been incorporated into the genome
such as retroviral sequences. CpG dinucleotides also exist in so-called
CpG islands, stretches of DNA characterized by a high
CG content, which are found in the majority of human
gene promoters. CpG islands in promoter regions are
typically unmethylated, and the lack of methylation
facilitates transcription.
Histone methylation involves the addition of a
methyl group to lysine residues in histone proteins
(Fig. 466-7). Depending on the specific lysine residue
being methylated, this alters chromatin configuration,
making it either more open or tightly packed. Acetylation of histone proteins is another well-characterized
mechanism that results in an open chromatin configuration, which favors active transcription. Acetylation
is generally more dynamic than methylation, and
many transcriptional activation complexes have histone acetylase activity, whereas repressor complexes
often contain deacetylases and remove acetyl groups
from histones. Other histone modifications include,
among others, phosphorylation and sumoylation.
Furthermore, noncoding RNAs and RNA regulatory networks that bind to DNA have a significant
impact on transcriptional activity.
Physiologically, epigenetic mechanisms play an
important role in several instances. For example,
X-inactivation refers to the relative silencing of one of
the two X chromosome copies present in females. The
inactivation process is a form of dosage compensation
such that females (XX) do not generally express twice
as many X-chromosomal gene products as males
(XY). In a given cell, the choice of which chromosome
is inactivated occurs randomly in humans. But once
the maternal or paternal X chromosome is inactivated,
Principles of Human Genetics
3645CHAPTER 466
it will remain inactive, and this information is transmitted with each
cell division. The X-inactive specific transcript (Xist) gene encodes a
large noncoding RNA that mediates the silencing of the X chromosome
from which it is transcribed by coating it with Xist RNA. The inactive
X chromosome is highly methylated and has low levels of histone acetylation. While the majority of X-chromosomal genes are silenced by
X-inactivation, ~15% escape inactivation and are expressed.
Epigenetic gene inactivation also occurs on selected chromosomal
regions of autosomes, a phenomenon referred to as genomic imprinting.
Through this mechanism, a small subset of genes is only expressed in
a monoallelic fashion. Imprinting is heritable and leads to the preferential expression of one of the parental alleles, which deviates from the
usual biallelic expression seen for the majority of genes. Remarkably,
imprinting can be limited to a subset of tissues. Imprinting is mediated through DNA methylation of one of the alleles. The epigenetic
marks on imprinted genes are maintained throughout life, but during
zygote formation, they are activated or inactivated in a sex-specific
manner (imprint reset) (Fig. 466-8), which allows a differential
expression pattern in the fertilized egg and the subsequent mitotic
divisions. Appropriate expression of imprinted genes is important for
normal development and cellular functions. Imprinting defects and
uniparental disomy, which is the inheritance of two chromosomes or
chromosomal regions from the same parent, are the cause of several
developmental disorders such as Beckwith-Wiedemann syndrome,
Silver-Russell syndrome, Angelman’s syndrome, and Prader-Willi
syndrome (see below). Monoallelic loss-of-function mutations in the
GNAS1 gene lead to Albright’s hereditary osteodystrophy (AHO).
Paternal transmission of GNAS1 mutations leads to an isolated AHO
phenotype (pseudopseudohypoparathyroidism), whereas maternal
transmission leads to AHO in combination with hormone resistance
to parathyroid hormone, thyrotropin, and gonadotropins (pseudohypoparathyroidism type IA). These phenotypic differences are explained
by tissue-specific imprinting of the GNAS1 gene, which is expressed
primarily from the maternal allele in the thyroid, gonadotropes, and
the proximal renal tubule. In most other tissues, the GNAS1 gene is
expressed biallelically. In patients with isolated renal resistance to
parathyroid hormone (pseudohypoparathyroidism type IB), defective
imprinting of the GNAS1 gene results in decreased Gs
α expression in
the proximal renal tubules. Rett’s syndrome is an X-linked dominant
disorder resulting in developmental regression and stereotypic hand
movements in affected girls. It is caused by mutations in the MECP2
gene, which encodes a methyl-binding protein. The ensuing aberrant
methylation results in abnormal gene expression in neurons, which are
otherwise normally developed.
Remarkably, epigenetic differences also occur among monozygotic
twins. Although twins are epigenetically indistinguishable during the
early years of life, older monozygotic twins exhibit differences in the
overall content and genomic distribution of DNA methylation and
histone acetylation, which would be expected to alter gene expression
in various tissues.
In cancer, the epigenome is characterized by simultaneous losses
and gains of DNA methylation in different genomic regions, as well
as repressive histone modifications. Hyper- and hypomethylation are
associated with mutations in genes that control DNA methylation.
Hypomethylation is thought to remove normal control mechanisms
that prevent expression of repressed DNA regions. It is also associated
with genomic instability. Hypermethylation, in contrast, results in
the silencing of CpG islands in promoter regions of genes, including
tumor-suppressor genes. Epigenetic alterations are considered to be
more easily reversible compared to genetic changes; modification of
the epigenome with demethylating agents and histone deacetylases is
being used in the treatment of various malignancies.
■ TRANSMISSION OF GENETIC DISEASE
Origins and Types of Mutations The term mutation or variant
is used to designate the process of generating genetic variations as well
as the outcome of these alterations. A mutation can be defined as any
change in the primary nucleotide sequence of DNA regardless of its
functional consequences, although it often has a negative connotation.
The more neutral term variation is now increasingly used to describe
sequence changes and is recommended by several professional organizations and guidelines instead of mutation. Some variations may be
lethal, others are less deleterious, and some may confer an evolutionary
advantage. Variations can occur in the germline (sperm or oocytes);
these can be transmitted to progeny. Alternatively, variations can occur
during embryogenesis or in somatic tissues. Variations that occur
during development lead to mosaicism, a situation in which tissues are
composed of cells with different genetic constitutions. If the germline
is mosaic, a mutation can be transmitted to some progeny but not
others, which sometimes leads to confusion in assessing the pattern
of inheritance. Somatic mutations that do not affect cell survival can
sometimes be detected because of variable phenotypic effects in tissues (e.g., pigmented lesions in McCune-Albright syndrome). Other
somatic mutations are associated with neoplasia because they confer
a growth advantage to cells. Epigenetic events may also influence gene
expression or facilitate genetic damage. With the exception of triplet
TABLE 466-2 Selected Examples of Diseases Caused by Mutations and
Rearrangements in Transcription Factors
TRANSCRIPTION
FACTOR CLASS EXAMPLE ASSOCIATED DISORDER
Nuclear receptors Androgen
receptor
Complete or partial androgen
insensitivity (recessive missense
mutations)
Spinobulbar muscular atrophy (CAG
repeat expansion)
Zinc finger proteins WT1 WAGR syndrome: Wilms’
tumor, aniridia, genitourinary
malformations, mental retardation
Basic helix-loop-helix MITF Waardenburg’s syndrome type 2A
Homeobox IPF1 Maturity onset of diabetes mellitus
type 4 (heterozygous mutation/
haploinsufficiency)
Pancreatic agenesis (homozygous
mutation)
Leucine zipper Retina leucine
zipper (NRL)
Autosomal dominant retinitis
pigmentosa
High mobility group
(HMG) proteins
SRY Sex reversal
Forkhead HNF4α, HNF1α,
HNF1β
Maturity onset of diabetes mellitus
types 1, 3, 5
Paired box PAX3 Waardenburg’s syndrome types 1
and 3
T-box TBX5 Holt-Oram syndrome (thumb
anomalies, atrial or ventricular
septum defects, phocomelia)
Cell cycle control
proteins
P53 Li-Fraumeni syndrome, other
cancers
Co-activators CREB binding
protein (CREBBP)
Rubinstein-Taybi syndrome
General transcription
factors
TATA-binding
protein (TBP)
Spinocerebellar ataxia 17 (CAG
expansion)
Transcription
elongation factor
VHL von Hippel–Lindau syndrome
(renal cell carcinoma,
pheochromocytoma, pancreatic
tumors, hemangioblastomas)
Autosomal dominant inheritance,
somatic inactivation of second allele
(Knudson two-hit model)
Runt RUNX1 Familial thrombocytopenia with
propensity to acute myelogenous
leukemia
Chimeric proteins due
to translocations
PML-RAR Acute promyelocytic leukemia
t(15;17)(q22;q11.2-q12) translocation
Abbreviations: CREB, cAMP responsive element–binding protein; HNF, hepatocyte
nuclear factor; PML, promyelocytic leukemia; RAR, retinoic acid receptor; SRY,
sex-determining region Y; VHL, von Hippel–Lindau.
3646 PART 16 Genes, the Environment, and Disease
Methylated DNA
Methylation
Histone Acetylation
Unmethylated DNA
Transcription
NH2
O
N
N
NH2
O
N
N
CH3
Histone Modifications
Cytosine Methylation
Acetylation
Phosphorylation
Methylation
NH2
FIGURE 466-7 Epigenetic modifications of DNA and histones. Methylation of cytosine residues is associated with
gene silencing. Methylation of certain genomic regions is inherited (imprinting), and it is involved in the silencing of
one of the two X chromosomes in females (X-inactivation). Alterations in methylation can also be acquired, e.g., in
cancer cells. Covalent posttranslational modifications of histones play an important role in altering DNA accessibility
and chromatin structure and hence in regulating transcription. Histones can be reversibly modified in their aminoterminal tails, which protrude from the nucleosome core particle, by acetylation of lysine, phosphorylation of serine,
methylation of lysine and arginine residues, and sumoylation. Acetylation of histones by histone acetylases (HATs),
e.g., leads to unwinding of chromatin and accessibility to transcription factors. Conversely, deacetylation by histone
deacetylases (HDACs) results in a compact chromatin structure and silencing of transcription.
nucleotide repeats, which can expand (see below), variations are usually stable.
Mutations are structurally diverse—they can involve the entire
genome, as in triploidy (one extra set of chromosomes), or gross
numerical or structural alterations in chromosomes or individual
genes. Large deletions may affect a portion of a gene or an entire gene,
or, if several genes are involved, they may lead to a contiguous gene syndrome. Unequal crossing-over between homologous genes can result
in fusion gene mutations, as illustrated by color blindness. Variations
involving single nucleotides are referred to as point mutations. Substitutions are called transitions if a purine is replaced by another purine base
(A ↔ G) or if a pyrimidine is replaced by another pyrimidine (C ↔ T).
Changes from a purine to a pyrimidine, or vice versa, are referred to as
transversions. If the DNA sequence change occurs in a coding region
and alters an amino acid, it is called a missense mutation. Depending
on the functional consequences of such a missense mutation, amino
acid substitutions in different regions of the protein can lead to distinct
phenotypes.
Variations can occur in all domains of a gene (Fig. 466-9). A point
mutation occurring within the coding region leads to an amino acid
substitution if the codon is altered (Fig. 466-10). Point mutations that
introduce a premature stop codon result in a truncated or missing
protein. Large deletions may affect a portion of a gene or an entire
gene, whereas small deletions and insertions alter the reading frame
if they do not represent a multiple of three bases. These “frameshift”
mutations, also designated as amphigoric amino acid changes, lead to
an entirely altered carboxy terminus. Mutations in intronic sequences
or in exon junctions may destroy or create splice donor or splice acceptor sites. Variations may also be found in the regulatory sequences of
genes, resulting in reduced or enhanced gene transcription.
Certain DNA sequences are particularly susceptible to mutagenesis.
Successive pyrimidine residues (e.g., T-T or C-C) are subject to the
formation of ultraviolet light–induced photoadducts. If these pyrimidine dimers are not repaired by the nucleotide excision repair pathway,
mutations will be introduced after DNA synthesis. The dinucleotide
C-G, or CpG, is also a hot spot for a specific type of mutation. In this
case, methylation of the cytosine is associated with an enhanced rate
of deamination to uracil, which is then replaced with thymine. This
C → T transition (or G → A on the opposite
strand) accounts for at least one-third of
point mutations associated with polymorphisms and mutations. In addition to the
fact that certain types of mutations (C →
T or G → A) are relatively common, the
nature of the genetic code also results in
overrepresentation of certain amino acid
substitutions.
Polymorphisms are sequence variations
that have a frequency of at least 1%. Usually,
they do not result in a perceptible phenotype,
but because allele frequency and functional
consequences are often not known, the term
variation is now increasingly recommended
for the description of these sequence changes.
Often, they consist of single base-pair substitutions that do not alter the protein coding
sequence because of the degenerate nature
of the genetic code (synonymous polymorphism), although it is possible that some
might alter mRNA stability, translation, or
the amino acid sequence (nonsynonymous
polymorphism) (Fig. 466-10). The detection
of sequence variants poses a practical problem because it is often unclear whether it creates a change with functional consequences
or a benign variation. In this situation, the
sequence alteration is also described as variant of unknown significance (VUS).
MUTATION RATES Mutations represent an important cause of genetic
diversity as well as disease. Mutation rates are difficult to determine
in humans because many mutations are silent and because testing is
often not adequate to detect the phenotypic consequences. Mutation
rates vary in different genes but are estimated to occur at a rate of
~10−10/bp per cell division. Germline mutation rates (as opposed
to somatic mutations) are relevant in the transmission of genetic
disease. Because the population of oocytes is established very early
in development, only ~20 cell divisions are required for completed
oogenesis, whereas spermatogenesis involves ~30 divisions by the time
of puberty and 20 cell divisions each year thereafter. Consequently,
the probability of acquiring new point mutations is much greater in
the male germline than the female germline, in which rates of aneuploidy are increased. Thus, the incidence of new point mutations in
spermatogonia increases with paternal age (e.g., achondrodysplasia,
Marfan’s syndrome, neurofibromatosis). It is estimated that about 1
in 10 sperm carries a new deleterious mutation. The rates for new
mutations are calculated most readily for autosomal dominant and
X-linked disorders and are ~10−5−10−6/locus per generation. Because
most monogenic diseases are relatively rare, new mutations account
for a significant fraction of cases. This is important in the context of
genetic counseling, because a new mutation can be transmitted to the
affected individual but does not necessarily imply that the parents are
at risk to transmit the disease to other children. An exception to this is
when the new mutation occurs early in germline development, leading
to gonadal mosaicism.
UNEQUAL CROSSING-OVER Normally, DNA recombination in germ
cells occurs with remarkable fidelity to maintain the precise junction
sites for the exchanged DNA sequences (Fig. 466-6). However, mispairing of homologous sequences leads to unequal crossover, with gene
duplication on one of the chromosomes and gene deletion on the other
chromosome. A significant fraction of growth hormone (GH) gene deletions, for example, involve unequal crossing-over (Chap. 379). The GH
gene is a member of a large gene cluster that includes a GH variant gene
as well as several structurally related chorionic somatomammotropin
genes and pseudogenes (highly homologous but functionally inactive
relatives of a normal gene). Because such gene clusters contain multiple
homologous DNA sequences arranged in tandem, they are particularly
Principles of Human Genetics
3647CHAPTER
Active
466
unmethylated
mat pat
Inactive
methylated
Maternal somatic cell
Inactive
methylated
pat mat
Active
unmethylated
Paternal somatic cell
Inactive
methylated
pat mat
Active
unmethylated
Zygote
Active
unmethylated
mat pat
Active
unmethylated
Maternal germline Paternal germline
Inactive
methylated
pat mat
Inactive
methylated
Germline development:
Imprint reset
FIGURE 466-8 A few genomic regions are imprinted in a parent-specific fashion. The unmethylated chromosomal
regions are actively expressed, whereas the methylated regions are silenced. In the germline, the imprint is reset
in a parent-specific fashion: both chromosomes are unmethylated in the maternal (mat) germline and methylated
in the paternal (pat) germline. In the zygote, the resulting imprinting pattern is identical with the pattern in the
somatic cells of the parents.
prone to undergo recombination and, consequently, gene duplication
or deletion. On the other hand, duplication of the PMP22 gene because
of unequal crossing-over results in increased gene dosage and type IA
Charcot-Marie-Tooth disease. Unequal crossing-over resulting in deletion of PMP22 causes a distinct neuropathy called hereditary neuropathy
with liability to pressure palsies (HNPP) (Chap. 446).
Glucocorticoid-remediable aldosteronism (GRA) is caused by a
gene fusion or rearrangement involving the genes that encode aldosterone synthase (CYP11B2) and steroid 11β-hydroxylase (CYP11B1),
normally arranged in tandem on chromosome 8q. These two genes
are 95% identical, predisposing to gene duplication and deletion by
unequal crossing-over. The rearranged gene product contains the
regulatory regions of 11β-hydroxylase fused to the coding sequence of
aldosterone synthetase. Consequently, the latter enzyme is expressed in
the adrenocorticotropic hormone (ACTH)–dependent zona fasciculata
of the adrenal gland, resulting in overproduction of mineralocorticoids
and hypertension (Chap. 386).
Gene conversion refers to a nonreciprocal exchange of homologous
genetic information. It has been used to explain how an internal
portion of a gene is replaced by a homologous segment copied from another allele or
locus; these genetic alterations may range
from a few nucleotides to a few thousand
nucleotides. As a result of gene conversion,
it is possible for short DNA segments of two
chromosomes to be identical, even though
these sequences are distinct in the parents. A
practical consequence of this phenomenon is
that nucleotide substitutions can occur during gene conversion between related genes,
often altering the function of the gene. In
disease states, gene conversion often involves
intergenic exchange of DNA between a gene
and a related pseudogene. For example, the
21-hydroxylase gene (CYP21A2) is adjacent
to a nonfunctional pseudogene (CYP21A1P).
Many of the nucleotide substitutions that are
found in the CYP21A2 gene in patients with
congenital adrenal hyperplasia correspond to
sequences that are present in the CYP21A1P
pseudogene, suggesting gene conversion as
one cause of mutagenesis. In addition, mitotic
gene conversion has been suggested as a
mechanism to explain revertant mosaicism
in which an inherited mutation is “corrected”
in certain cells. For example, patients with
autosomal recessive generalized atrophic
benign epidermolysis bullosa have acquired
reverse mutations in one of the two mutated
COL17A1 alleles, leading to clinically unaffected patches of skin.
INSERTIONS AND DELETIONS Although
many instances of insertions and deletions
occur as a consequence of unequal crossingover, there is also evidence for internal
duplication, inversion, or deletion of DNA
sequences. The fact that certain deletions or
insertions appear to occur repeatedly as independent events indicates that specific regions
within the DNA sequence predispose to these
errors. For example, certain regions of the
DMD gene, which encodes dystrophin, appear
to be hot spots for deletions and result in muscular dystrophy (Chap. 449). Some regions
within the human genome are rearrangement
hot spots and lead to CNVs.
ERRORS IN DNA REPAIR Because mutations
caused by defects in DNA repair accumulate
as somatic cells divide, these types of mutations are particularly important in the context of neoplastic disorders. Several genetic disorders
involving DNA repair enzymes underscore their importance. Patients
with xeroderma pigmentosum have defects in DNA damage recognition or in the nucleotide excision and repair pathway (Chap. 76).
Exposed skin is dry and pigmented and is extraordinarily sensitive to
the mutagenic effects of ultraviolet irradiation. More than 10 different
genes have been shown to cause the different forms of xeroderma
pigmentosum.
Ataxia-telangiectasia is a multisystem disorder that includes progressive neurodegenerative cerebellar ataxia, immunologic defects, telangiectatic lesions, lymphomas and leukemias, and hypersensitivity to
ionizing radiation (Chap. 439). The discovery of the ataxia-telangiectasia
mutated (ATM) gene revealed that it is homologous to genes involved
in DNA repair and control of cell cycle checkpoints. Mutations in
the ATM gene give rise to defects in meiosis as well as increasing
susceptibility to damage from ionizing radiation. Fanconi’s anemia
is also associated with an increased risk of multiple acquired genetic
abnormalities. It is characterized by diverse congenital anomalies and
No comments:
Post a Comment
اكتب تعليق حول الموضوع