Sei sulla pagina 1di 30

CMDT 2015 861

21
Electrolyte & Acid-Base
Disorders

Kerry C. Cho, MD

ASSESSMENT OF THE PATIENT fractional excretion (Fe) of an electrolyte X (Fex) calculated


from a spot urine sample:
The diagnosis and treatment of fluid and electrolyte disor-
ders are based on (1) careful history, (2) physical examina-
Urine X/Serum X
tion and assessment of total body water and its distribution, F E x (%) =   × 100
(3) serum electrolyte concentrations, (4) urine electrolyte Urine Cr/Serum Cr
concentrations, and (5) serum osmolality. The patho-
physiology of electrolyte disorders is rooted in basic prin- A low fractional excretion indicates renal reabsorption
ciples of total body water and its distribution across fluid (high avidity or electrolyte retention), while a high frac-
compartments. tional excretion indicates renal wasting (low avidity or
electrolyte excretion). Thus, the fractional excretion helps
the clinician determine whether the kidney’s response is
A. Body Water and Fluid Distribution appropriate for a specific electrolyte disorder.
Total body water is different in men than in women, and it
decreases with aging (Table 21–1). Approximately 50–60% D. Serum Osmolality
of total body weight is water; two-thirds (40% of body Solute concentration is measured by osmolality in milli-
weight) is intracellular, while one-third (20% of body moles per kilogram. Osmolarity is measured in millimoles
weight) is extracellular. One-fourth of extracellular fluid of solute per liter of solution. At physiologic solute concen-
(5% of body weight) is intravascular. Water may be lost trations (normally 285–295 mmol/kg), the two measure-
from either or both compartments (intracellular and extra- ments are clinically interchangeable. Tonicity refers to
cellular). Changes in total body water content are best osmolytes that are impermeable to cell membranes. Differ-
evaluated by documenting changes in body weight. Effec- ences in osmolyte concentration across cell membranes
tive circulating volume may be assessed by physical exami- lead to osmosis and fluid shifts, stimulation of thirst, and
nation (eg, blood pressure, pulse, jugular venous secretion of antidiuretic hormone (ADH). Substances that
distention). Quantitative measurements of effective circu- easily permeate cell membranes (eg, urea, ethanol) are
lating volume and intravascular volume may be invasive ineffective osmoles that do not cause fluid shifts across
(ie, central venous pressure or pulmonary wedge pressure) fluid compartments.
or noninvasive (ie, inferior vena cava diameter and right Serum osmolality can be estimated using the following
atrial pressure by echocardiography) but still require care- formula:
ful interpretation.
Osmolality =
B. Serum Electrolytes Glucose mg/dL BUN mg/dL
2(Na+ mEq/L) + +
The cause of electrolyte disorders may be determined by 18 2.8
reviewing the history, underlying diseases, and medications.
(1 mosm/L of glucose equals 180 mg/L or 18 mg/dL and 1
mosm/L of urea nitrogen equals 28 mg/L or 2.8 mg/dL).
C. Evaluation of Urine
Sodium is the major extracellular cation; doubling the
The urine concentration of an electrolyte indicates renal serum sodium in the formula for estimated osmolality
handling of the electrolyte and whether the kidney is accounts for counterbalancing anions. A discrepancy
appropriately excreting or retaining the electrolyte. A between measured and estimated osmolality of  > 10 mmol/
24-hour urine collection for daily electrolyte excretion is kg suggests an osmolal gap, which is the presence of
the gold standard for renal electrolyte handling, but it is unmeasured osmoles such as ethanol, methanol, isopropa-
slow and onerous. A more convenient method is the nol, and ethylene glycol (see Table 38–5).

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 861 7/11/14 5:23 PM
862 CMDT 2015 Chapter 21

▶▶General Considerations
Table 21–1.  Total body water (as percentage of body
weight) in relation to age and sex. Defined as a serum sodium concentration < 135 mEq/L (or
< 135 mmol/L), hyponatremia is the most common electro-
Age Male Female lyte abnormality in hospitalized patients. The clinician
18–40 60% 50% should be wary about hyponatremia since mismanagement
can result in neurologic catastrophes from cerebral osmotic
41–60 60–50% 50–40% demyelination. Indeed, iatrogenic complications from
Over 60 50% 40% aggressive or inappropriate therapy can be more harmful
than hyponatremia itself.
A common misconception is that the sodium concen-
▼▼
DISORDERS OF SODIUM CONCENTRATION tration is a reflection of total body sodium or total body
water. In fact, total body water and sodium can be low,
HYPONATREMIA normal, or high in hyponatremia since the kidney inde-
pendently regulates sodium and water homeostasis. Most
cases of hyponatremia reflect water imbalance and abnor-
ESSENTIALS OF DIAGNOSIS mal water handling, not sodium imbalance, indicating the
primary role of ADH in the pathophysiology of hyponatre-
mia. A diagnostic algorithm using serum osmolality and
▶▶ Volume status and serum osmolality are essential volume status separates the causes of hyponatremia into
to determine etiology. therapeutically useful categories (Figure 21–1).
▶▶ Hyponatremia usually reflects excess water reten-
tion relative to sodium rather than sodium defi- ▶▶Etiology
ciency. The sodium concentration is not a measure
A. Isotonic & Hypertonic Hyponatremia
of total body sodium.
▶▶ Hypotonic fluids commonly cause hyponatremia Serum osmolality identifies isotonic and hypertonic hypo-
in hospitalized patients. natremia, although these cases can often be identified by
careful history or previous laboratory tests.

HYPONATREMIA

Serum osmolality

Normal Low High


(280–295 mosm/kg) (< 280 mosm/kg) (> 295 mosm/kg)

Isotonic Hypotonic Hypertonic


hyponatremia hyponatremia hyponatremia
1. Hyperproteinemia 1. Hyperglycemia
2. Hyperlipidemia (chylomicrons, 2. Mannitol, sorbitol,
triglycerides, rarely cholesterol) glycerol, maltose
3. Radiocontrast agents
Volume status

Hypovolemic Euvolemic Hypervolemic

UNa+ < 10 mEq/L UNa+ > 20 mEq/L 1. SIADH Edematous states


Extrarenal salt loss Renal salt loss 2. Postoperative hyponatremia 1. Heart failure
1. Dehydration 1. Diuretics 3. Hypothyroidism 2. Liver disease
2. Diarrhea 2. ACE inhibitors 4. Psychogenic polydipsia 3. Nephrotic syndrome
3. Vomiting 3. Nephropathies 5. Beer potomania (rare)
4. Mineralocorticoid 6. Idiosyncratic drug reaction (thiazide 4. Advanced kidney
deficiency diuretics, ACE inhibitors) disease
5. Cerebral sodium- 7. Endurance exercise
wasting syndrome 8. Adrenocorticotropin deficiency

▲▲Figure 21–1.  Evaluation of hyponatremia using serum osmolality and extracellular fluid volume status. ACE, angioten-
sin-converting enzyme; SIADH, syndrome of inappropriate antidiuretic hormone. (Adapted, with permission, from Narins
RG et al. Diagnostic strategies in disorders of fluid, electrolyte and acid-base homeostasis. Am J Med. 1982 Mar;72(3):496–520.)

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 862 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 863

Isotonic hyponatremia is seen with severe hyperlipid- includes renal sodium wasting possibly through B-type
emia and hyperproteinemia. Lipids (including chylomi- natriuretic peptide, ADH release, and decreased aldoste-
crons, triglycerides, and cholesterol) and proteins (> 10 g/dL rone secretion.
[> 100 g/L], eg, paraproteinemias and intravenous immuno-
2. Euvolemic hypotonic hyponatremia—Euvolemic hypo-
globulin therapy) interfere with the measurement of serum
natremia has the broadest differential diagnosis. Most
sodium, causing pseudohyponatremia. Serum osmolality is
causes are mediated directly or indirectly through ADH,
isotonic because lipids and proteins do not affect osmolality
including hypothyroidism, adrenal insufficiency, medica-
measurement. Newer sodium assays using ion-specific elec-
tions, and the syndrome of inappropriate ADH (SIADH).
trodes on undiluted serum specimens (ie, the direct assay
The exceptions are primary polydipsia, beer potomania,
method) will not result in pseudohyponatremia.
and reset osmostat.
Hypertonic hyponatremia occurs with hyperglycemia
and mannitol administration for increased intracranial A. hormonal abnormalities—Hypothyroidism and
pressure. Glucose and mannitol osmotically pull intracel- adrenal insufficiency can cause hyponatremia. Exactly how
lular water into the extracellular space. The translocation hypothyroidism induces hyponatremia is unclear but may
of water lowers the serum sodium concentration. Translo- be related to ADH. Adrenal insufficiency may be associ-
cational hyponatremia is not pseudohyponatremia or an ated with the hyperkalemia and metabolic acidosis of
artifact of sodium measurement. The sodium concentra- hypoaldosteronism. Cortisol provides feedback inhibition
tion falls 2 mEq/L (or 2 mmol/L) for every 100 mg/dL (or for ADH release.
5.56 mmol/L) rise in glucose when the glucose concentra-
B. Thiazide diuretics and other medications—
tion is between 200 mg/dL and 400 mg/dL (11.1 mmol/L
Thiazides induce hyponatremia typically in older female
and 22.2 mmol/L). If the glucose concentration is > 400
patients within days of initiating therapy. The mechanism
mg/dL, the sodium concentration falls 4 mEq/L for every
appears to be a combination of mild diuretic-induced vol-
100 mg/dL rise in glucose. There is some controversy about
ume contraction, ADH effect, and intact urinary concen-
the correction factor for the serum sodium in the presence
trating ability resulting in water retention and hyponatremia.
of hyperglycemia. Many guidelines recommend a correc-
Loop diuretics do not cause hyponatremia as frequently
tion factor, whereby the serum sodium concentration
because of disrupted medullary concentrating gradient and
decreases by 1.6 mEq/L (or 1.6 mmol/L) for every 100 mg/
impaired urine concentration.
dL (5.56 mmol/L) rise in plasma glucose above normal, but
Nonsteroidal anti-inflammatory drugs (NSAIDs)
there is evidence that the decrease may be greater when
increase ADH by inhibiting prostaglandin formation. Pros-
patients have more severe hyperglycemia (> 400 mg/dL or
taglandins and selective serotonin reuptake inhibitors (eg,
22.2 mmol/L) or volume depletion, or both. One group has
fluoxetine, paroxetine, and citalopram) can cause hypona-
suggested (based on short-term exposure of normal volun-
tremia, especially in geriatric patients. Enhanced secretion
teers to markedly elevated glucose levels) that when the
or action of ADH may result from increased serotonergic
serum glucose is > 200 mg/dL, the serum sodium concen-
tone. Angiotensin-converting enzyme (ACE) inhibitors do
tration decreases by at least 2.4 mEq/L (or 2.4 mmol/L).
not block the conversion of angiotensin I to angiotensin II
in the brain. Angiotensin II stimulates thirst and ADH
B. Hypotonic Hyponatremia secretion. Hyponatremia during amiodarone loading has
Most cases of hyponatremia are hypotonic, highlighting been reported; it usually improves with dose reduction.
sodium’s role as the predominant extracellular osmole. The Abuse of 3,4-methylenedioxymethamphetamine (MDMA,
next step is classifying hypotonic cases by the patient’s vol- also known as Ecstasy) can lead to hyponatremia and severe
ume status. neurologic symptoms, including seizures, cerebral edema,
and brainstem herniation. MDMA and its metabolites
1. Hypovolemic hypotonic hyponatremia—Hypovolemic increase ADH release from the hypothalamus. Primary poly-
hyponatremia occurs with renal or extrarenal volume loss dipsia may contribute to hyponatremia since MDMA users
and hypotonic fluid replacement (Figure 21–1). Total body typically increase fluid intake to prevent hyperthermia.
sodium and total body water are decreased. To maintain
C. Nausea, pain, surgery, and medical proce-
intravascular volume, the pituitary increases ADH secre-
dures—Nausea and pain are potent stimulators of ADH
tion, causing free water retention from hypotonic fluid
release. Severe hyponatremia can develop after elective
replacement. The body sacrifices serum osmolality to pre-
surgery in healthy patients, especially premenopausal
serve intravascular volume. In short, losses of water and
women. Hypotonic fluids in the setting of elevated ADH
salt are replaced by water alone. Without ongoing hypo-
levels can produce severe, life-threatening hyponatremia.
tonic fluid intake, the renal or extrarenal volume loss
Medical procedures such as colonoscopy have also been
would produce hypovolemic hypernatremia.
associated with hyponatremia.
Cerebral salt wasting is a distinct and rare subset of
hypovolemic hyponatremia seen in patients with intracra- D. HIV infection—Hyponatremia is seen in up to 50% of
nial disease (eg, infections, cerebrovascular accidents, hospitalized HIV-infected patients and 20% of ambulatory
tumors, and neurosurgery). Clinical features include HIV-infected patients. The differential diagnosis is broad:
refractory hypovolemia and hypotension, often requiring medication effect, adrenal insufficiency, hypoaldosteron-
continuous infusion of isotonic or hypertonic saline and ism, central nervous system or pulmonary disease, SIADH,
ICU monitoring. The exact pathophysiology is unclear but malignancy, and volume depletion.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 863 7/11/14 5:23 PM
864 CMDT 2015 Chapter 21

E. Exercise-associated hyponatremia—Hyponatre-
mia after exercise, especially endurance events such as tri- Table 21–2.  Causes of syndrome of inappropriate ADH
athlons and marathons, may be caused by a combination of secretion (SIADH).
excessive hypotonic fluid intake and continued ADH Central nervous system disorders
secretion. Reperfusion of the exercise-induced ischemic   Head trauma
splanchnic bed causes delayed absorption of excessive  Stroke
quantities of hypotonic fluid ingested during exercise. Sus-   Subarachnoid hemorrhage
tained elevation of ADH prevents water excretion in this  Hydrocephalus
setting. Current guidelines suggest that endurance athletes   Brain tumor
drink water according to thirst rather than according to  Encephalitis
specified hourly rates of fluid intake. Specific universal   Guillain-Barré syndrome
 Meningitis
recommendations for fluid replacement rates are not pos-
  Acute psychosis
sible given the variability of sweat production, renal water   Acute intermittent porphyria
excretion, and environmental conditions. Electrolyte- Pulmonary lesions
containing sport drinks do not protect against hyponatre-  Tuberculosis
mia since they are markedly hypotonic relative to serum.   Bacterial pneumonia
 Aspergillosis
F. Syndrome of inappropriate antidiuretic hor-  Bronchiectasis
mone secretion—Under normal circumstances, hypovo-  Neoplasms
lemia and hyperosmolality stimulate ADH secretion. ADH   Positive pressure ventilation
release is inappropriate without these physiologic cues. Malignancies
Normal regulation of ADH release occurs from both the   Bronchogenic carcinoma
central nervous system and the chest via baroreceptors and   Pancreatic carcinoma
neural input. The major causes of SIADH (Table 21–2) are   Prostatic carcinoma
  Renal cell carcinoma
disorders affecting the central nervous system (structural,
  Adenocarcinoma of colon
metabolic, psychiatric, or pharmacologic processes) or the  Thymoma
lungs (infectious, mechanical, oncologic). Medications  Osteosarcoma
commonly cause SIADH by increasing ADH or its action.  Lymphoma
Some carcinomas, especially small cell lung carcinoma, can  Leukemia
autonomously secrete ADH. Drugs
  Increased ADH production
G. Psychogenic polydipsia and beer potomania—    Antidepressants: tricyclics, monoamine oxidase inhibitors,
Marked free water intake (generally > 10 L/d) may produce   SSRIs
hyponatremia. Euvolemia is maintained through renal    Antineoplastics: cyclophosphamide, vincristine
excretion of sodium. Urine sodium is therefore generally   Carbamazepine
elevated (> 20 mEq/L), and ADH levels are appropriately    Methylenedioxymethamphetamine (MDMA; Ecstasy)
suppressed. As the increased free water is excreted, the   Clofibrate
urine osmolality approaches the minimum of 50 mosm/kg    Neuroleptics: thiothixene, thioridazine, fluphenazine,
  haloperidol, trifluoperazine
(or 50 mmol/kg). Polydipsia occurs in psychiatric patients.
  Potentiated ADH action
Psychiatric medications may interfere with water excretion   Carbamazepine
or increase thirst through anticholinergic side effects, fur-   Chlorpropamide, tolbutamide
ther increasing water intake. The hyponatremia of beer   Cyclophosphamide
potomania occurs in patients who consume large amounts   NSAIDs
of beer. Free water excretion is decreased because of    Somatostatin and analogs
decreased solute consumption and production; muscle   Amiodarone
wasting and malnutrition are contributing factors. Without Others
enough solute, these patients have decreased free water  Postoperative
 Pain
excretory capacity even if they maximally dilute the urine.
 Stress
H. Reset osmostat—Reset osmostat is a rare cause of hypo-  AIDS
natremia characterized by appropriate ADH regulation in   Pregnancy (physiologic)
 Hypokalemia
response to water deprivation and fluid challenges. Patients
with reset osmostat regulate serum sodium and serum osmo- ADH, antidiuretic hormone; NSAIDs, nonsteroidal anti-inflamma-
lality around a lower set point, concentrating or diluting urine tory drugs; SSRIs, selective serotonin reuptake inhibitors.
in response to hyperosmolality and hypo-osmolality. The mild
hypo-osmolality of pregnancy is a form of reset osmostat.
vasodilation or decreased cardiac output. Increased renin-
3. Hypervolemic hypotonic hyponatremia—Hypervol- angiotensin-aldosterone system activity and ADH secre-
emic hyponatremia occurs in the edematous states of cir- tion result in water retention. Note the pathophysiologic
rhosis, heart failure, nephrotic syndrome, and advanced similarity to hypovolemic hyponatremia—the body sacri-
kidney disease (Figure 21–1). In cirrhosis and heart failure, fices osmolality in an attempt to restore effective circulating
effective circulating volume is decreased due to peripheral volume.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 864 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 865

The pathophysiology of hyponatremia in nephrotic result from increased urea and uric acid clearances in
syndrome is not completely understood, but the primary response to the volume-expanded state. Azotemia may
disturbance may be renal sodium retention, resulting in reflect volume contraction, ruling out SIADH, which is
overfilling of the intravascular space and secondary edema seen in euvolemic patients.
formation as fluid enters the interstitial space. Previously, it
was thought that the decreased oncotic pressure of hypoal- ▶▶Complications
buminemia caused fluid shifts from the intravascular space
The most serious complication of hyponatremia is iatro-
to the interstitial compartment. Intravascular underfilling
genic cerebral osmotic demyelination from overly rapid
led to secondary renal sodium retention. However, patients
sodium correction. Also called central pontine myelinoly-
receiving therapy for glomerular disease and nephrotic
sis, cerebral osmotic demyelination may occur outside the
syndrome often have edema resolution prior to normaliza-
brainstem. Demyelination may occur days after sodium
tion of the serum albumin.
correction or initial neurologic recovery from hyponatre-
Patients with advanced kidney disease typically have
mia. Hypoxic episodes during hyponatremia may contrib-
sodium retention and decreased free water excretory
ute to demyelination. The neurologic effects are generally
capacity, resulting in hypervolemic hyponatremia.
catastrophic and irreversible.

▶▶Clinical Findings ▶▶Treatment


A. Symptoms and Signs Regardless of the patient’s volume status, another common
Whether hyponatremia is symptomatic depends on its feature is to restrict free water and hypotonic fluid intake,
severity and acuity. Chronic disease can be severe (sodium since these solutions will exacerbate hyponatremia. Free
concentration < 110 mEq/L), yet remarkably asymptomatic water intake from oral intake and intravenous fluids should
because the brain has adapted by decreasing its tonicity over generally be < 1–1.5 L/d.
weeks to months. Acute disease that has developed over Hypovolemic patients require adequate fluid resuscita-
hours to days can be severely symptomatic with relatively tion from isotonic fluids (either normal saline or lactated
modest hyponatremia. Mild hyponatremia (sodium con- Ringer solution) to suppress the hypovolemic stimulus for
centrations of 130–135 mEq/L) is usually asymptomatic. ADH release. Patients with cerebral salt wasting may
Mild symptoms of nausea and malaise progress to head- require hypertonic saline to prevent circulatory collapse;
ache, lethargy, and disorientation as the sodium concentra- some may respond to fludrocortisone. Hypervolemic
tion drops. The most serious symptoms are respiratory patients may require loop diuretics or dialysis, or both, to
arrest, seizure, coma, permanent brain damage, brainstem correct increased total body water and sodium. Euvolemic
herniation, and death. Premenopausal women are much patients may respond to free water restriction alone.
more likely than menopausal women to die or suffer per- Pseudohyponatremia from hypertriglyceridemia or
manent brain injury from hyponatremic encephalopathy, hyperproteinemia requires no therapy except confirmation
suggesting a hormonal role in the pathophysiology. with the clinical laboratory. Translocational hyponatre-
Evaluation starts with a careful history for new medica- mia from glucose or mannitol can be managed with glu-
tions, changes in fluid intake (polydipsia, anorexia, intrave- cose correction or mannitol discontinuation (if possible).
nous fluid rates and composition), fluid output (nausea No specific therapy is necessary in patients with reset
and vomiting, diarrhea, ostomy output, polyuria, oliguria, osmostat since they successfully regulate their serum
insensible losses). The physical examination should help sodium with fluid challenges and water deprivation.
categorize the patient’s volume status into hypovolemia, Symptomatic and severe hyponatremia generally
euvolemia, or hypervolemia. require hospitalization for careful monitoring of fluid bal-
ance and weights, treatment, and frequent sodium checks.
Inciting medications should be discontinued if possible.
B. Laboratory Findings
There is no consensus about the optimal rate of sodium
Laboratory assessment should include serum electrolytes, correction in symptomatic hyponatremic patients. Recent
creatinine, and osmolality as well as urine sodium. The guidelines have introduced new recommendations. First, a
etiology of most cases of hyponatremia will be apparent relatively small increase of 4–6 mEq/L in the serum sodium
from the history, physical, and basic laboratory tests. Addi- may be all that is necessary to reverse the neurologic mani-
tional tests of thyroid and adrenal function will occasion- festations of symptomatic hyponatremia. Second, acute
ally be necessary. hyponatremia (eg, exercise-associated hyponatremia) with
SIADH is a clinical diagnosis characterized by (1) hypo- severe neurologic manifestations can be reversed rapidly
natremia; (2) decreased osmolality (< 280 mosm/kg [< 280 with 100 mL of 3% hypertonic saline infused over 10 min-
mmol/kg]); (3) absence of heart, kidney, or liver disease; utes (repeated twice as necessary). Third, lower correction
(4) normal thyroid and adrenal function (see Chapter 26); rates for chronic hyponatremia have been introduced, as
and (5) urine sodium usually over 20 mEq/L. In clinical low as 4–8 mEq/L per 24 hours in patients at high risk for
practice, ADH levels are not measured. Patients with demyelination. Fourth, chronic hyponatremic patients at
SIADH may have low blood urea nitrogen (BUN) (< 10 high risk for demyelination who are corrected too rapidly
mg/dL [or < 3.6 mmol/L]) and hypouricemia (< 4 mg/dL are candidates for treatment with a combination of DDAVP
[or < 238 mcmol/L]), which are not only dilutional but and intravenous dextrose 5% to relower the serum sodium.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 865 7/11/14 5:23 PM
866 CMDT 2015 Chapter 21

In severely symptomatic patients, the clinician should for patients receiving vasopressin antagonists since the
calculate the sodium deficit and deliver 3% hypertonic aquaresis can result in excessive sodium correction in a
saline. The sodium deficit can be calculated by the follow- fluid-restricted patient. Frequent monitoring of the serum
ing formula: sodium is necessary.

Sodium deficit = Total body water (TBW) ▶▶When to Refer


          × (Desired serum Na–Actual serum Na) • Nephrology or endocrinology consultation should be
considered in severe, symptomatic, refractory, or com-
where TBW is typically 50% of total mass in women and plicated cases of hyponatremia.
55% of total mass in men. For example, a nonedematous,
• Aggressive therapies with hypertonic saline, demeclo-
severely symptomatic 70 kg woman with a serum sodium
cycline, vasopressin antagonists, or dialysis mandate
of 124 mEq/L should have her serum sodium corrected to
specialist consultation.
approximately 132 mEq/L in the first 24 hours. Her sodium
deficit is calculated as: • Consultation may be necessary with end-stage liver or
heart disease.
Sodium deficit = 70 kg × 0.5 × (132 mEq/L − 124 mEq/L)
= 280 mEq ▶▶When to Admit
3% hypertonic saline has a sodium concentration of 514 Hospital admission is necessary for symptomatic patients
mEq/1000 mL. The delivery rate for hypertonic saline can or those requiring aggressive therapies for close monitor-
be calculated as: ing and frequent laboratory testing.

Delivery rate = Sodium deficit/(514 mEq/1000mL)/


24 hours Graff-Radford J et al. Clinical and radiologic correlations of
central pontine myelinolysis syndrome. Mayo Clin Proc. 2011
= 280 mEq/(514 mEq/1000 mL)/24 hours
Nov;86(11):1063–7. [PMID: 21997578]
= 22 mL/hour Lehrich RW et al. Role of vaptans in the management of hypona-
tremia. Am J Kidney Dis. 2013 Aug;62(2):364–76. [PMID:
In general, the 3% hypertonic saline infusion rate should 23725974]
not exceed 0.5 mL/kg body weight/h; higher rates may Leung AA et al. Preoperative hyponatremia and perioperative
represent a miscalculated sodium deficit or a mathematical complications. Arch Intern Med. 2012 Oct 22;172(19):
error. Hypertonic saline in hypervolemic patients can be 1474–81. [PMID: 22965221]
hazardous, resulting in worsening volume overload, pul- Pokaharel M et al. Dysnatremia in the ICU. Curr Opin Crit Care.
monary edema, and ascites. 2011 Dec;17(6):581–93. [PMID: 22027406]
Shchekochikhin D et al. Hyponatremia: an update on current
For patients who cannot adequately restrict free water pharmacotherapy. Expert Opin Pharmacother. 2013
or have an inadequate response to conservative measures, Apr;14(6):747–55. [PMID: 23496346]
demeclocycline (300–600 mg orally twice daily) inhibits Verbalis JG et al. Diagnosis, evaluation, and treatment of hypo-
the effect of ADH on the distal tubule. Onset of action may natremia: expert panel recommendations. Am J Med. 2013
require 1 week, and urinary concentrating ability may be Oct;126(10 Suppl 1):S1–42. [PMID: 24074529]
permanently impaired, resulting in nephrogenic diabetes
insipidus (DI) and even hypernatremia. Cirrhosis may
increase the nephrotoxicity of demeclocycline. HYPERNATREMIA
Vasopressin antagonists may revolutionize the treat-
ment of euvolemic and hypervolemic hyponatremia, espe-
cially in heart failure. Tolvaptan, lixivaptan, and satavaptan ESSENTIALS OF DIAGNOSIS
are oral selective vasopressin-2 receptor antagonists;
conivaptan is an intravenous agent. Tolvaptan and conivap- ▶▶ Increased thirst and water intake is the first
tan are available in the United States, but lixivaptan and defense against hypernatremia.
satavaptan are not yet approved by the US Food and Drug
Administration (FDA).
▶▶ Urine osmolality helps differentiate renal from
V2 receptors mediate the diuretic effect of ADH and V2 nonrenal water loss.
receptor antagonists are recommended for use in hospital.
For hospitalized patients with euvolemic SIADH, tolvaptan
▶▶General Considerations
is begun as 15 mg orally daily and can be increased to
30 mg daily and 60 mg daily at 24 hour intervals if hypona- Hypernatremia is defined as a sodium concentration > 145
tremia persists or if the increase in sodium concentration is mEq/L. All patients with hypernatremia have hyperosmo-
< 5 mEq/L over the preceding 24 hours. Conivaptan is lality, unlike hyponatremic patients who can have a low,
given as an intravenous loading dose of 20 mg delivered normal, or high serum osmolality. The hypernatremic
over 30 minutes, then as 20 mg continuously over 24 hours. patient is typically hypovolemic due to free water losses,
Subsequent infusions may be administered every 1–3 days although hypervolemia is frequently seen, often as an iat-
at 20–40 mg/d by continuous infusion. The standard free rogenic complication in hospitalized patients with impaired
water restriction for hyponatremic patients should be lifted access to free water. Rarely, excessive sodium intake may

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 866 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 867

cause hypernatremia. Hypernatremia in primary aldoste- (1 mmol/L/h). There is no consensus about the optimal rates
ronism is mild and usually does not cause symptoms. of sodium correction in hypernatremia and hyponatremia.
An intact thirst mechanism and access to water are the
primary defense against hypernatremia. The hypothalamus A. Choice of Type of Fluid for Replacement
can sense minimal changes in serum osmolality, triggering
the thirst mechanism and increased water intake. Thus, 1. Hypernatremia with hypovolemia—Hypovolemic
whatever the underlying disorder (eg, dehydration, lactu- patients should receive isotonic 0.9% normal saline to
lose or mannitol therapy, central and nephrogenic DI), restore euvolemia and to treat hyperosmolality because
excess water loss can cause hypernatremia only when ade- normal saline (308 mosm/kg or 308 mmol/kg) is hypo-
quate water intake is not possible. osmolar compared with plasma. After adequate volume
resuscitation with normal saline, 0.45% saline or 5% dex-
trose (or both) can be used to replace any remaining free
▶▶Clinical Findings water deficit. Milder volume deficits may be treated with
A. Symptoms and Signs 0.45% saline and 5% dextrose.

When the patient is dehydrated, orthostatic hypotension 2. Hypernatremia with euvolemia—Water ingestion or
and oliguria are typical findings. Because water shifts from intravenous 5% dextrose will result in the excretion of
the cells to the intravascular space to protect volume status, excess sodium in the urine. If the glomerular filtration rate
these symptoms may be delayed. Lethargy, irritability, and (GFR) is decreased, diuretics will increase urinary sodium
weakness are early signs. Hyperthermia, delirium, seizures, excretion but may impair renal concentrating ability,
and coma may be seen with severe hypernatremia (ie, increasing the quantity of water that needs to be replaced.
sodium > 158 mEq/L). Symptoms in the elderly may not be 3. Hypernatremia with hypervolemia—Treatment
specific; a recent change in consciousness is associated with includes 5% dextrose solution to reduce hyperosmolality.
a poor prognosis. Osmotic demyelination is an uncommon Loop diuretics may be necessary to promote natriuresis
but reported consequence of severe hypernatremia. and lower total body sodium. In severe rare cases with
kidney disease, hemodialysis may be necessary to correct
B. Laboratory Findings the excess total body sodium and water.
1. Urine osmolality > 400 mosm/kg—Renal water-
conserving ability is functioning. B. Calculation of Water Deficit
A. Nonrenal losses—Hypernatremia will develop if Fluid replacement should include the free water deficit and
water intake falls behind hypotonic fluid losses from exces- additional maintenance fluid to replace ongoing and antic-
sive sweating, the respiratory tract, or bowel movements. ipated fluid losses.
Lactulose causes an osmotic diarrhea with loss of free 1. Acute hypernatremia—In acute dehydration without
water. much solute loss, free water loss is similar to the weight
B. Renal losses—While severe hyperglycemia can cause loss. Initially, a 5% dextrose solution may be used. As cor-
translocational hyponatremia, progressive volume deple- rection of water deficit progresses, therapy should continue
tion from glucosuria can result in hypernatremia. Osmotic with 0.45% saline with dextrose.
diuresis can occur with the use of mannitol or urea. 2. Chronic hypernatremia—The water deficit is calculated
2. Urine osmolality < 250 mosm/kg—Hypernatremia with to restore normal sodium concentration, typically 140
a dilute urine (osmolality < 250 mosm/kg) is characteristic mEq/L. Total body water (TBW) (Table 21–1) correlates
of DI. Central DI results from inadequate ADH release. with muscle mass and therefore decreases with advancing
Nephrogenic DI results from renal insensitivity to ADH; age, cachexia, and dehydration and is lower in women than
common causes include lithium, demeclocycline, relief of in men. Current TBW equals 40–60% current body weight.
urinary obstruction, interstitial nephritis, hypercalcemia,
and hypokalemia. Volume (in L) = Current TBW ×   [Na+] − 140
to be repleced 140
▶▶Treatment
Treatment of hypernatremia includes correcting the cause ▶▶When to Refer
of the fluid loss, replacing water, and replacing electrolytes Patients with refractory or unexplained hypernatremia
(as needed). In response to increases in plasma osmolality, should be referred for subspecialist consultation.
brain cells synthesize solutes called idiogenic osmoles,
which cause intracellular fluid shifts. Osmole production ▶▶When to Admit
begins 4–6 hours after dehydration and takes several days
to reach steady state. If hypernatremia is rapidly cor- • Patients with symptomatic hypernatremia require hos-
rected, the osmotic imbalance may cause cerebral edema pitalization for evaluation and treatment.
and potentially severe neurologic impairment. Fluids • Significant comorbidities or concomitant acute ill-
should be administered over a 48-hour period, aiming for nesses, especially if contributing to hypernatremia, may
serum sodium correction of approximately 1 mEq/L/h necessitate hospitalization.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 867 7/11/14 5:23 PM
868 CMDT 2015 Chapter 21

increased anion gap (see Chapter 38). The combination of


Al-Absi A et al. A clinical approach to the treatment of chronic
hypernatremia. Am J Kidney Dis. 2012 Dec;60(6):1032–8.
an increased anion gap metabolic acidosis and an osmol
[PMID: 22959761] gap exceeding 10 mosm/kg (or 10 mmol/kg) is not specific
Alshayeb HM et al. Severe hypernatremia correction rate and for toxic alcohol ingestion and may occur with alcoholic
mortality in hospitalized patients. Am J Med Sci. 2011 ketoacidosis or lactic acidosis (see Metabolic Acidosis).
May;341(5):356–60. [PMID: 21358313]
Arampatzis S et al. Characteristics, symptoms, and outcome of Kruse JA. Methanol and ethylene glycol intoxication. Crit Care
severe dysnatremias present on hospital admission. Am J Clin. 2012 Oct;28(4):661–711. [PMID: 22998995]
Med. 2012 Nov;125(11):1125.e1–e7. [PMID: 22939097]
Lindner G et al. Hypernatremia in critically ill patients. J Crit
Care. 2013 Apr;28(2):216.e11–20. [PMID: 22762930] HYPEROSMOLALITY ASSOCIATED WITH
Sam R et al. Understanding hypernatremia. Am J Nephrol. SIGNIFICANT SHIFTS IN WATER
2012;36(1):97–104. [PMID: 22739333]
Increased concentrations of solutes that do not readily enter
cells cause a shift of water from intracellular to extracellular.
VOLUME OVERLOAD
Hyperosmolality of effective osmoles such as sodium and
glucose causes symptoms, primarily neurologic. The sever-
ESSENTIALS OF DIAGNOSIS ity of symptoms depends on the degree of hyperosmolality
and rapidity of development. In acute hyperosmolality,
somnolence and confusion can appear when the osmolality
▶▶ Disorder of excessive sodium retention in the set- exceeds 320–330 mosm/kg (320–330 mmol/kg); coma,
ting of low arterial underfilling (eg, heart failure or respiratory arrest, and death can result when osmolality
cirrhosis). exceeds 340–350 mosm/kg (340–350 mmol/kg).
▶▶ Hyponatremia from water retention in edematous
states is associated with sodium retention. Kraut JA et al. Approach to the evaluation of a patient with an
increased serum osmolal gap and high-anion-gap metabolic
acidosis. Am J Kidney Dis. 2011 Sep;58(3):480–4. [PMID:
21794966]
The hallmark of a volume overloaded state is sodium reten-
Whittington JE et al. The osmolal gap: what has changed? Clin
tion. Abnormally low arterial filling, such as from heart Chem. 2010 Aug;56(8):1353–5. [PMID: 20530730]
failure or cirrhosis, activates the neurohumoral axis, which
stimulates the renin-angiotensin-aldosterone system, the ▼▼
sympathetic nervous system, and ADH (vasopressin) DISORDERS OF POTASSIUM
release. The result is sodium retention with edema. The CONCENTRATION
stimulus for vasopressin release is nonosmotic. Released in
response to baroreceptor activation, vasopressin stimulates HYPOKALEMIA
renal V2 receptors, resulting in water reabsorption, edema
formation, and hyponatremia.
ESSENTIALS OF DIAGNOSIS
Bagshaw SM et al. Disorders of sodium and water balance in
hospitalized patients. Can J Anaesth. 2009 Feb;56(2):151–67. ▶▶ Serum potassium level < 3.5 mEq/L (< 3.5 mmol/L)
[PMID: 19247764]
Rosner MH et al. Dysnatremias in the intensive care unit. Con- ▶▶ Severe hypokalemia may induce dangerous
trib Nephrol. 2010;165:292–8. [PMID: 20427980] arrhythmias and rhabdomyolysis.
▶▶ Transtubular potassium concentration gradient
▼▼ (TTKG) can distinguish renal from nonrenal loss of
HYPEROSMOLAR DISORDERS & potassium.
OSMOLAR GAPS
HYPEROSMOLALITY WITH TRANSIENT OR NO ▶▶General Considerations
SIGNIFICANT SHIFT IN WATER
Hypokalemia can result from insufficient dietary potas-
Urea and alcohol readily cross cell membranes and can sium intake, intracellular shifting of potassium from the
produce hyperosmolality. Urea is an ineffective osmole extracellular space, extrarenal potassium loss, or renal
with little effect on osmotic water movement across cell potassium loss (Table 21–3). Cellular uptake of potassium
membranes. Alcohol quickly equilibrates between the is increased by insulin and beta-adrenergic stimulation
intracellular and extracellular compartments, adding 22 and blocked by alpha-adrenergic stimulation. Aldosterone
mosm/L for every 100 mg/dL (or 21.7 mmol/L) of ethanol. is an important regulator of total body potassium, increas-
Ethanol ingestion should be considered in any case of stu- ing potassium secretion in the distal renal tubule. The
por or coma with an elevated osmol gap (measured osmo- most common cause of hypokalemia, especially in devel-
lality – calculated osmolality > 10 mosm/kg [> 10 mmol/ oping countries, is gastrointestinal loss from infectious
kg]). Other toxic alcohols such as methanol and ethylene diarrhea. The potassium concentration in intestinal secretion
glycol cause an osmol gap and a metabolic acidosis with an is ten times higher (80 mEq/L) than in gastric secretions.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 868 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 869

Table 21–3.  Causes of hypokalemia. Table 21–4.  Genetic disorders associated with electro-
lyte metabolism disturbances.
Decreased potassium intake
Potassium shift into the cell Disease Site of Mutation
  Increased postprandial secretion of insulin
 Alkalosis Potassium
  Trauma (via beta-adrenergic stimulation?)
 Hypokalemia
  Periodic paralysis (hypokalemic)
  Barium intoxication   Hypokalemic periodic Dihydropyridine-sensitive skeletal
Renal potassium loss   paralysis muscle voltage-gated calcium
  Increased aldosterone (mineralocorticoid) effects channel
  Primary hyperaldosteronism   Bartter syndrome Na+-K+-2Cl– cotransporter, K+
   Secondary aldosteronism (dehydration, heart failure) channel (ROMK), or Cl– channel
  Renovascular hypertension of thick ascending limb of Henle
  Malignant hypertension (hypofunction), barttin
   Ectopic ACTH-producing tumor
  Gitelman syndrome   Gitelman syndrome Thiazide-sensitive
  Bartter syndrome Na+-Cl– cotransporter
  Cushing syndrome   Liddle syndrome Beta or gamma subunit of
  Licorice (European) amiloride-sensitive Na+ channel
  Renin-producing tumor (hyperfunction)
   Congenital abnormality of steroid metabolism (eg,
  Apparent mineralo- 11-beta-hydroxysteroid dehydro-
   adrenogenital syndrome, 17-alpha-hydroxylase defect,
  corticoid excess genase (failure to inactivate
   apparent mineralocorticoid excess, 11-beta-hydroxylase
cortisol)
  deficiency)
  Increased flow to distal nephron   Glucocorticoid- Regulatory sequence of 11-beta-
   Diuretics (furosemide, thiazides)   remediable hydroxysteroid controls aldoste-
  Salt-losing nephropathy   hyperaldosteronism rone synthase inappropriately
 Hypomagnesemia  Hyperkalemia
  Unreabsorbable anion
  Carbenicillin, penicillin   Hyperkalemic periodic Alpha subunit of calcium channel
  Renal tubular acidosis (type I or II)    paralysis
  Fanconi syndrome   Pseudohypoaldoste- Beta or gamma subunit of
  Interstitial nephritis    ronism type I amiloride-sensitive Na+ channel
   Metabolic alkalosis (bicarbonaturia) (hypofunction)
  Congenital defect of distal nephron
  Liddle syndrome   Pseudohypoaldoster- HNK2, HNK4
Extrarenal potassium loss    onism type II
  Vomiting, diarrhea, laxative abuse   (Gordon syndrome)
  Villous adenoma, Zollinger-Ellison syndrome Calcium
  Familial hypocalciuric Ca2+-sensing protein
 hypercalcemia (hypofunction)
Hypokalemia in the presence of acidosis suggests profound   Familial hypocalcemia Ca2+-sensing protein
potassium depletion and requires urgent treatment. Self- (hyperfunction)
limited hypokalemia occurs in 50–60% of trauma patients, Phosphate
perhaps related to enhanced release of epinephrine.
 Hypophosphatemic PEX gene, FGF23
Hypokalemia increases the likelihood of digitalis toxic-  rickets
ity. In patients with heart disease, hypokalemia induced by
beta-2-adrenergic agonists and diuretics may substantially Magnesium
increase the risk of arrhythmias. Numerous genetic muta-  Hypomagnesemia- Paracellin-1
tions affect fluid and electrolyte metabolism, including  hypercalciuria
disorders of potassium metabolism (Table 21–4).  syndrome
Magnesium is an important cofactor for potassium Water
uptake and maintenance of intracellular potassium levels.   Nephrogenic diabetes Vasopressin receptor-2 (Type 1),
Loop diuretics (eg, furosemide) cause substantial renal  insipidus aquaporin-2
potassium and magnesium losses. Magnesium depletion
Acid-base
should be considered in refractory hypokalemia.
  Proximal RTA Na+ HCO3– cotransporter
▶▶Clinical Findings   Distal RTA Cl– HCO3– exchanger H+-ATPase
A. Symptoms and Signs   Proximal and distal RTA Carbonic anhydrase II

Muscular weakness, fatigue, and muscle cramps are fre- FGF23, fibroblast growth factor 23; RTA, renal tubular acidosis.
quent complaints in mild to moderate hypokalemia.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 869 7/11/14 5:23 PM
870 CMDT 2015 Chapter 21

Gastrointestinal smooth muscle involvement may result in ▶▶When to Refer


constipation or ileus. Flaccid paralysis, hyporeflexia, hyper-
capnia, tetany, and rhabdomyolysis may be seen with Patients with unexplained hypokalemia, refractory hyper-
severe hypokalemia (< 2.5 mEq/L). The presence of hyper- kalemia, or clinical features suggesting alternative diagno-
tension may be a clue to the diagnosis of hypokalemia from ses (eg, aldosteronism or hypokalemic periodic paralysis)
aldosterone or mineralocorticoid excess (Table 21–4). should be referred for endocrinology or nephrology
Renal manifestations include nephrogenic DI and intersti- consultation.
tial nephritis.
▶▶When to Admit
B. Laboratory Findings Patients with symptomatic or severe hypokalemia, especially
Urinary potassium concentration is low (< 20 mEq/L) as a with cardiac manifestations, require cardiac monitoring,
result of extrarenal loss (eg, diarrhea, vomiting) and inap- frequent laboratory testing, and potassium supplementation.
propriately high (> 40 mEq/L) with renal loss (eg, miner-
alocorticoid excess, Bartter syndrome, Liddle syndrome) Asmar A et al. A physiologic-based approach to the treatment
(Table 21–3). of a patient with hypokalemia. Am J Kidney Dis. 2012
The transtubular [K+] gradient (TTKG) is a simple and Sep;60(3):492–7. [PMID: 22901631]
Marti G et al. Etiology and symptoms of severe hypokalemia in
rapid evaluation of net potassium secretion. TTKG is cal-
emergency department patients. Eur J Emerg Med. 2014
culated as follows: Feb;21(1):46–51. [PMID: 23839104]
Pepin J et al. Advances in diagnosis and management of hypoka-
Urine K + /Plasma K + lemic and hyperkalemic emergencies. Emerg Med Pract. 2012
TTKG =   Feb;14(2):1–18. [PMID: 22413702]
Urine osm/Plasma osm Rastegar A. Attending rounds: patient with hypokalemia and
metabolic acidosis. Clin J Am Soc Nephrol. 2011 Oct;6(10):
Hypokalemia with a TTKG > 4 suggests renal potassium 2516–21. [PMID: 21921151]
loss with increased distal K+ secretion. In such cases,
plasma renin and aldosterone levels are helpful in differen-
tial diagnosis. The presence of nonabsorbed anions, such HYPERKALEMIA
as bicarbonate, increases the TTKG.

C. Electrocardiogram ESSENTIALS OF DIAGNOSIS


The electrocardiogram (ECG) shows decreased amplitude
and broadening of T waves, prominent U waves, premature ▶▶ Serum potassium level > 5.0 mEq/L (> 5.0 mmol/L).
ventricular contractions, and depressed ST segments. ▶▶ Hyperkalemia may develop in patients taking ACE
inhibitors, angiotensin-receptor blockers, potas-
sium-sparing diuretics, or their combination, even
▶▶Treatment with no or only mild kidney dysfunction.
Oral potassium supplementation is the safest and easiest ▶▶ The ECG may show peaked T waves, widened QRS
treatment for mild to moderate deficiency. Dietary potas- and biphasic QRS–T complexes, or may be normal
sium is almost entirely coupled to phosphate—rather than despite life-threatening hyperkalemia.
chloride—and is therefore not effective in correcting ▶▶ Measurement of plasma potassium level differen-
potassium loss associated with chloride depletion from tiates potassium leak from blood cells in cases of
diuretics or vomiting. In the setting of abnormal kidney clotting, leukocytosis, and thrombocytosis from
function and mild to moderate diuretic dosage, 20 mEq/d elevated serum potassium.
of oral potassium is generally sufficient to prevent hypo-
kalemia, but 40–100 mEq/d over a period of days to weeks
▶▶ Rule out extracellular potassium shift from the cells
is needed to treat hypokalemia and fully replete potassium in acidosis and assess renal potassium excretion.
stores.
Intravenous potassium is indicated for patients with
▶▶General Considerations
severe hypokalemia and for those who cannot take oral
supplementation. For severe deficiency, potassium may be Hyperkalemia usually occurs in patients with advanced
given through a peripheral intravenous line in a concentra- kidney disease but can also develop with normal kidney
tion up to 40 mEq/L and at rates up to 10 mEq/h. Concen- function (Table 21–5). Acidosis causes intracellular potas-
trations of up to 20 mEq/h may be given through a central sium to shift extracellularly. Serum potassium concentra-
venous catheter. Continuous ECG monitoring is indicated, tion rises about 0.7 mEq/L for every decrease of 0.1 pH
and the serum potassium level should be checked every unit during acidosis. Fist clenching during venipuncture
3–6 hours. Avoid glucose-containing fluid to prevent fur- may raise the potassium concentration by 1–2 mEq/L by
ther shifts of potassium into the cells. Magnesium defi- causing acidosis and potassium shift from cells. In the
ciency should be corrected, particularly in refractory absence of acidosis, serum potassium concentration rises
hypokalemia. about 1 mEq/L when there is a total body potassium excess

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 870 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 871

Trimethoprim is structurally similar to amiloride and


Table 21–5.  Causes of hyperkalemia. triamterene, and all three drugs inhibit renal potassium
Spurious/Pseudohyperkalemia excretion through suppression of sodium channels in the
  Leakage from erythrocytes when separation of serum from clot distal nephron.
  is delayed (plasma K+ normal) Cyclosporine and tacrolimus can induce hyperkalemia
  Marked thrombocytosis or leukocytosis with release of in organ transplant recipients, especially kidney transplant
  intracellular K+ (plasma K+ normal) patients, partly due to suppression of the basolateral
  Repeated fist clenching during phlebotomy, with release of K+ Na+–K+-ATPase in principal cells. Hyperkalemia is com-
  from forearm muscles monly seen in HIV patients and has been attributed to
  Specimen drawn from arm with intravenous K+ infusion impaired renal excretion of potassium due to pentamidine
Decreased K+ excretion
or trimethoprim-sulfamethoxazole or to hyporeninemic
  Kidney disease, acute and chronic
  Renal secretory defects (may or may not have reduced kidney hypoaldosteronism.
  function): kidney transplant, interstitial nephritis, systemic
  lupus erythematosus, sickle cell disease, amyloidosis, ▶▶Clinical Findings
  obstructive nephropathy
  Hyporeninemic hypoaldosteronism (often in diabetic patients Hyperkalemia impairs neuromuscular transmission, caus-
  with mild to moderate nephropathy) or selective ing muscle weakness, flaccid paralysis, and ileus. Electro-
  hypoaldosteronism (eg, AIDS patients) cardiography is not a sensitive method for detecting
  Drugs that inhibit potassium excretion: spironolactone, hyperkalemia, since nearly half of patients with a serum
  eplerenone, drospirenone, NSAIDs, ACE inhibitors, angiotensin potassium level > 6.5 mEq/L will not manifest ECG changes.
  II receptor blockers, triamterene, amiloride, trimethoprim,
ECG changes in hyperkalemia include bradycardia, PR
  pentamidine, cyclosporine, tacrolimus
Shift of K+ from within the cell
interval prolongation, peaked T waves, QRS widening, and
  Massive release of intracellular K+ in burns, rhabdomyolysis, biphasic QRS–T complexes. Conduction disturbances,
  hemolysis, severe infection, internal bleeding, vigorous such as bundle branch block and atrioventricular block,
 exercise may occur. Ventricular fibrillation and cardiac arrest are
  Metabolic acidosis (in the case of organic acid accumulation— terminal events.
  eg, lactic acidosis—a shift of K+ does not occur since organic
  acid can easily move across the cell membrane)
  Hypertonicity (solvent drag)
▶▶Prevention
  Insulin deficiency (metabolic acidosis may not be apparent) Inhibitors of the renin-angiotensin-aldosterone axis (ie,
  Hyperkalemic periodic paralysis ACE inhibitors, ARBs, and spironolactone) and potas-
  Drugs: succinylcholine, arginine, digitalis toxicity, beta-adrenergic sium-sparing diuretics (eplerenone, triamterene) should be
 antagonists used cautiously in patients with heart failure, liver failure,
  Alpha-adrenergic stimulation?
and kidney disease. Laboratory monitoring should be per-
Excessive intake of K+
  Especially in patients taking medications that decrease potassium formed within 1 week of drug initiation or dosage increase.
  secretion (see above)
▶▶Treatment
ACE, angiotensin-converting enzyme; NSAIDs, nonsteroidal anti-
inflammatory drugs. The diagnosis should be confirmed by repeat laboratory
testing to rule out spurious hyperkalemia, especially in the
absence of medications that cause hyperkalemia or in
patients without kidney disease or a previous history of
of 1–4 mEq/kg. However, the higher the serum potassium hyperkalemia. Plasma potassium concentration can be
concentration, the smaller the excess necessary to raise the measured to avoid hyperkalemia due to potassium leakage
potassium levels further. out of red cells, white cells, and platelets. Kidney dysfunc-
Mineralocorticoid deficiency from Addison disease or tion should be ruled out at the initial assessment.
chronic kidney disease (CKD) is another cause of hyperka- Treatment consists of withholding exogenous potas-
lemia with decreased renal excretion of potassium. Miner- sium, identifying the cause, reviewing the patient’s medi-
alocorticoid resistance due to genetic disorders, interstitial cations and dietary potassium intake, and correcting the
kidney disease, or urinary tract obstruction also leads to hyperkalemia. Emergent treatment is indicated when car-
hyperkalemia. diac toxicity, muscle paralysis, or severe hyperkalemia
ACE inhibitors or angiotensin-receptor blockers (potassium > 6.5 mEq/L) is present, even in the absence of
(ARBs), commonly used in patients with heart failure or ECG changes. Insulin, bicarbonate, and beta-agonists shift
CKD, may cause hyperkalemia. The concomitant use of potassium intracellularly within minutes of administra-
spironolactone, eplerenone, or beta-blockers further tion (Table 21–6). Intravenous calcium may be given to
increases the risk of hyperkalemia. Thiazide or loop diuret- antagonize the cell membrane effects of potassium, but its
ics and sodium bicarbonate may minimize hyperkalemia. use should be restricted to life-threatening hyperkalemia
Persistent mild hyperkalemia in the absence of ACE inhibi- in patients taking digitalis because hypercalcemia may
tor or ARB therapy is usually due to type IV renal tubular cause digitalis toxicity. Hemodialysis may be required to
acidosis (RTA). Heparin inhibits aldosterone production in remove potassium in patients with acute or chronic kidney
the adrenal glands, causing hyperkalemia. injury.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 871 7/11/14 5:23 PM
872 CMDT 2015 Chapter 21

Table 21–6.  Treatment of hyperkalemia.

IMMEDIATE

Mechanism of K+ Removed
Modality Action Onset Duration Prescription from Body
Calcium Antagonizes car- 0–5 minutes 1 hour Calcium gluconate 10%, 5–30 mL 0
diac conduction intravenously; or calcium chloride 5%,
abnormalities 5–30 mL intravenously
Bicarbonate Distributes K+ into 15–30 minutes 1–2 hours NaHCO3, 44–88 mEq (1–2 ampules) 0
cells intravenously
Note: Sodium bicarbonate may not be effec-
tive in end-stage renal disease patients; dial-
ysis is more expedient and effective. Some
patients may not tolerate the additional
sodium load of bicarbonate therapy.
Insulin Distributes K+ into 15–60 minutes 4–6 hours Regular insulin, 5–10 units intravenously, plus 0
cells glucose 50%, 25 g intravenously
Albuterol Distributes K+ into 15–30 minutes 2–4 hours Nebulized albuterol, 10–20 mg in 4 mL normal 0
cells saline, inhaled over 10 minutes
Note: Much higher doses are necessary for
hyperkalemia therapy (10–20 mg) than for
airway disease (2.5 mg).
URGENT

Mechanism of K+ Removed
Modality Action Onset of Action Prescription from Body
Loop diuretic Renal K+ excretion 0.5–2 hours Furosemide, 40–160 mg intravenously Variable
Note: Diuretics may not be effective in patients
with acute and chronic kidney diseases.
Sodium polysty- Ion-exchange resin 1–3 hours Oral: 15–60 g in 20% sorbitol (60–240 mL) 0.5–1 mEq/g
rene sulfonate binds K+ Rectal: 30–60 g in 20% sorbitol resin
(eg, Kayexalate) Note: Resins with sorbitol may cause bowel
necrosis and intestinal perforation, especially
in postoperative patients.
Hemodialysis1 Extracorporeal K+ 1–8 hours Dialysate [K+] 0–1 mEq/L 25–50 mEq/h
removal Note: A fast and effective therapy for
hyperkalemia, hemodialysis can be delayed
by vascular access placement and equipment
and/or staffing availability. Serum K can be
rapidly corrected within minutes, but
post-dialysis rebound can occur.
Peritoneal dialysis Peritoneal K+ 1–4 hours Frequent exchanges 200–300 mEq
removal
1
Can be both acute immediate and urgent treatment of hyperkalemia.
Modified and reproduced, with permission, from Cogan MG. Fluid and Electrolytes: Physiology and Pathophysiology. McGraw-Hill, 1991.

▶▶When to Refer concomitant illness (eg, tumor lysis, rhabdomyolysis, met-


abolic acidosis) should be sent to the emergency depart-
• Patients with hyperkalemia from kidney disease and ment for immediate treatment.
reduced renal potassium excretion should see a
nephrologist. Kamel KS et al. Asking the question again: are cation exchange
• Transplant patients may need adjustment of their resins effective for the treatment of hyperkalemia? Nephrol
immunosuppression regimen by transplant specialists. Dial Transplant. 2012 Dec;27(12):4294–7. [PMID: 22989741]
Palmer BF. A physiologic-based approach to the evaluation of a
patient with hyperkalemia. Am J Kidney Dis. 2010
Aug;56(2):387–93. [PMID: 20493606]
▶▶When to Admit Pepin J et al. Advances in diagnosis and management of hypoka-
Patients with severe hyperkalemia > 6 mEq/L, any degree lemic and hyperkalemic emergencies. Emerg Med Pract. 2012
Feb;14(2):1–18. [PMID: 22413702]
of hyperkalemia associated with ECG changes, or

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 872 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 873

Shingarev R et al. A physiologic-based approach to the treatment Table 21–7.  Causes of hypocalcemia.
of acute hyperkalemia. Am J Kidney Dis. 2010 Sep;56(3):
578–84. [PMID: 20570423] Decreased intake or absorption
 Malabsorption
  Small bowel bypass, short bowel
▼▼   Vitamin D deficit (decreased absorption, decreased production
DISORDERS OF CALCIUM   of 25-hydroxyvitamin D or 1,25-dihydroxyvitamin D)
CONCENTRATION Increased loss
The normal total plasma (or serum) calcium concentration  Alcoholism
  Chronic kidney disease
is 8.5–10.5 mg/dL (or 2.1–2.6 mmol/L). Ionized calcium
  Diuretic therapy
(normal: 4.6–5.3 mg/dL [or 1.15–1.32 mmol/L]) is physi- Endocrine disease
ologically active and necessary for muscle contraction and   Hypoparathyroidism (genetic, acquired; including hypomagne-
nerve function.   semia and hypermagnesemia)
The calcium-sensing receptor, a transmembrane pro-   Post-parathyroidectomy (hungry bone syndrome)
tein that detects the extracellular calcium concentration,  Pseudohypoparathyroidism
has been identified in the parathyroid gland and the kid-   Calcitonin secretion with medullary carcinoma of the thyroid
ney. Functional defects in this protein are associated with   Familial hypocalcemia
diseases of abnormal calcium metabolism such as familial Associated diseases
hypocalcemia and familial hypocalciuric hypercalcemia  Pancreatitis
 Rhabdomyolysis
(Table 21–4).
  Septic shock
Physiologic causes
  Associated with decreased serum albumin1
HYPOCALCEMIA   Decreased end-organ response to vitamin D
 Hyperphosphatemia
  Induced by aminoglycoside antibiotics, plicamycin, loop diuretics,
ESSENTIALS OF DIAGNOSIS  foscarnet
1
Ionized calcium concentration is normal.
▶▶ Often mistaken as a neurologic disorder.
▶▶ Check for decreased serum parathyroid hormone
(PTH), vitamin D, or magnesium levels.
▶▶ If the ionized calcium level is normal despite a low ▶▶Clinical Findings
total serum calcium, calcium metabolism is usu-
ally normal. A. Symptoms and Signs
Hypocalcemia increases excitation of nerve and muscle
cells, primarily affecting the neuromuscular and cardiovas-
▶▶General Considerations
cular systems. Spasm of skeletal muscle causes cramps and
The most common cause of low total serum calcium is tetany. Laryngospasm with stridor can obstruct the airway.
hypoalbuminemia. When serum albumin concentration is Convulsions, perioral and peripheral paresthesias, and
lower than 4 g/dL (40 g/L), serum Ca2+ concentration is abdominal pain can develop. Classic physical findings
reduced by 0.8–1 mg/dL (0.20–0.25 mmol/L) for every 1 g/ include Chvostek sign (contraction of the facial muscle in
dL (10 g/L) of albumin. response to tapping the facial nerve) and Trousseau sign
The most accurate measurement of serum calcium is (carpal spasm occurring with occlusion of the brachial
the ionized calcium concentration. True hypocalcemia artery by a blood pressure cuff). QT prolongation predis-
(decreased ionized calcium) implies insufficient action of poses to ventricular arrhythmias. In chronic hypoparathy-
PTH or active vitamin D. Important causes of hypocalce- roidism, cataracts and calcification of basal ganglia may
mia are listed in Table 21–7. appear (see Chapter 26).
The most common cause of hypocalcemia is advanced
CKD, in which decreased production of active vitamin D3
B. Laboratory Findings
(1, 25 dihydroxyvitamin D3) and hyperphosphatemia both
play a role (see Chapter 22). Some cases of primary hypo- Serum calcium concentration is low (< 8.5 mg/dL [or < 2.1
parathyroidism are due to mutations of the calcium-sens- mmol/L]). In true hypocalcemia, the ionized serum cal-
ing receptor in which inappropriate suppression of PTH cium concentration is also low (< 4.6 mg/dL [or < 1.15
release leads to hypocalcemia (see Chapter 26). Magne- mmol/L]). Serum phosphate is usually elevated in hypo-
sium depletion reduces both PTH release and tissue parathyroidism or in advanced CKD, whereas it is sup-
responsiveness to PTH, causing hypocalcemia. Hypocalce- pressed in early CKD or vitamin D deficiency.
mia in pancreatitis is a marker of severe disease. Elderly Serum magnesium concentration is commonly low. In
hospitalized patients with hypocalcemia and hypophos- respiratory alkalosis, total serum calcium is normal but ion-
phatemia, with or without an elevated PTH level, are likely ized calcium is low. The ECG shows a prolonged QT
vitamin D deficient. interval.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 873 7/11/14 5:23 PM
874 CMDT 2015 Chapter 21

▶▶Treatment1 ▶▶ Most often, asymptomatic, mild hypercalcemia (≥


A. Severe, Symptomatic Hypocalcemia 10.5 mg/dL [or 2.6 mmol/L]) is due to primary
hyperparathyroidism, whereas the symptomatic,
In the presence of tetany, arrhythmias, or seizures, intravenous severe hypercalcemia (≥ 14 mg/dL [or 3.5 mmol/L])
calcium gluconate is indicated. Because of the short duration is due to hypercalcemia of malignancy.
of action, continuous calcium infusion is usually required.
Ten to 15 milligrams of calcium per kilogram body weight,
or six to eight 10-mL vials of 10% calcium gluconate ▶▶General Considerations
(558–744 mg of calcium), is added to 1 L of D5W and
infused over 4–6 hours. By monitoring the serum calcium Important causes of hypercalcemia are listed in Table 21–8.
level frequently (every 4–6 hours), the infusion rate is Primary hyperparathyroidism and malignancy account for
adjusted to maintain the serum calcium level at 7–8.5 mg/dL. 90% of cases. Primary hyperparathyroidism is the most
common cause of hypercalcemia (usually mild) in ambula-
B. Asymptomatic Hypocalcemia tory patients. Chronic hypercalcemia (over 6 months) or
some manifestation such as nephrolithiasis also suggests a
Oral calcium (1–2 g) and vitamin D preparations, including benign cause. Tumor production of PTH-related proteins
active vitamin D sterols, are used. Calcium carbonate is well (PTHrP) is the most common paraneoplastic endocrine
tolerated and less expensive than many other calcium tablets. syndrome, accounting for most cases of hypercalcemia in
A check of urinary calcium excretion is recommended after inpatients (see Table 39–2). The neoplasm is clinically
the initiation of therapy because hypercalciuria (urine cal- apparent in nearly all cases when the hypercalcemia is
cium excretion > 300 mg or > 7.5 mmol per day) or urine detected, and the prognosis is poor. Granulomatous dis-
calcium:creatinine ratio > 0.3 may impair kidney function in eases, such as sarcoidosis and tuberculosis, cause hypercal-
these patients. The low serum calcium associated with hypo- cemia via overproduction of active vitamin D3 (1,25
albuminemia does not require replacement therapy. If serum dihydroxyvitamin D3).
Mg2+ is low, therapy must include magnesium replacement, Milk-alkali syndrome has had a resurgence due to cal-
which by itself will usually correct hypocalcemia. cium ingestion for prevention of osteoporosis. Heavy cal-
cium carbonate intake causes hypercalcemic acute kidney
▶▶When to Refer injury, likely from renal vasoconstriction. The decreased
Patients with complicated hypocalcemia from hypopara- GFR impairs bicarbonate excretion, while hypercalcemia
thyroidism, familial hypocalcemia, or CKD require referral stimulates proton secretion and bicarbonate reabsorption.
to an endocrinologist or nephrologist. Metabolic alkalosis decreases calcium excretion, maintain-
ing hypercalcemia.
▶▶When to Admit
Patients with tetany, arrhythmias, seizures, or other symp-
toms of hypocalcemia require immediate evaluation and
Table 21–8.  Causes of hypercalcemia.
therapy.
Increased intake or absorption
Al-Azem H et al. Hypoparathyroidism. Best Pract Res Clin Endo-   Milk-alkali syndrome
crinol Metab. 2012 Aug;26(4):517–22. [PMID: 22863393]   Vitamin D or vitamin A excess
Fong J et al. Hypocalcemia: updates in diagnosis and manage- Endocrine disorders
ment for primary care. Can Fam Physician. 2012   Primary hyperparathyroidism
Feb;58(2):158–62. [PMID: 22439169]
  Secondary or tertiary hyperparathyroidism (usually associated
Kelly A et al. Hypocalcemia in the critically ill patient. J Intensive
  with hypocalcemia)
Care Med. 2013 May–Jun;28(3):166–77. [PMID: 21841146]
Peacock M. Calcium metabolism in health and disease. Clin J Am  Acromegaly
Soc Nephrol. 2010 Jan;5(Suppl 1):S23–30. [PMID: 20089499]   Adrenal insufficiency
 Pheochromocytoma
 Thyrotoxicosis
HYPERCALCEMIA Neoplastic diseases
  Tumors producing PTH-related proteins (ovary, kidney, lung)
  Multiple myeloma (elaboration of osteoclast-activating factor)
ESSENTIALS OF DIAGNOSIS   Lymphoma (occasionally from production of calcitriol)
Miscellaneous causes
  Thiazide diuretic use
▶▶ Primary hyperparathyroidism and malignancy-   Granulomatous diseases (production of calcitriol)
associated hypercalcemia are the most common   Paget disease of bone
causes.  Hypophosphatasia
 Immobilization
▶▶ Hypercalciuria usually precedes hypercalcemia.   Familial hypocalciuric hypercalcemia
  Complications of kidney transplantation
  Lithium intake
1
See also Chapter 26 for discussion of the treatment of
hypoparathyroidism. PTH, parathyroid hormone.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 874 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 875

Hypercalcemia causes nephrogenic DI through activa- 0.45% saline or 0.9% saline can be given rapidly (250–500
tion of calcium-sensing receptors in collecting ducts, mL/h). A meta-analysis questioned the efficacy and safety
which reduces ADH-induced water permeability. Volume profile of intravenous furosemide for hypercalcemia. Thia-
depletion further worsens hypercalcemia. zides can worsen hypercalcemia.
Bisphosphonates are the treatment of choice for hyper-
▶▶Clinical Findings calcemia of malignancy. Although they are safe, effective,
and normalize calcium in > 70% of patients, bisphospho-
A. Symptoms and Signs
nates may require up to 48–72 hours before reaching full
The history and physical examination should focus on the therapeutic effect. Calcitonin may be helpful in the short-
duration of hypercalcemia and evidence for a neoplasm. term until bisphosphonates reach therapeutic levels. In
Hypercalcemia may affect gastrointestinal, kidney, emergency cases, dialysis with low calcium dialysate may
and neurologic function. Mild hypercalcemia is often be needed. The calcimimetic agent cinacalcet hydrochlo-
asymptomatic. Symptoms usually occur if the serum cal- ride suppresses PTH secretion and decreases serum cal-
cium is > 12 mg/dL (or > 3 mmol/L) and tend to be more cium concentration and holds promise as a treatment
severe if hypercalcemia develops acutely. Symptoms option. (See Chapters 26 and 39.)
include constipation and polyuria, except in hypocalciuric Typically, if dialysis patients do not receive proper sup-
hypercalcemia, in which polyuria is absent. Other symp- plementation of calcium and active vitamin D, hypocalce-
toms include nausea, vomiting, anorexia, peptic ulcer dis- mia and hyperphosphatemia develop. On the other hand,
ease, renal colic, and hematuria from nephrolithiasis. hypercalcemia can sometimes develop, particularly in the
Polyuria from hypercalciuria-induced nephrogenic DI can setting of severe secondary hyperparathyroidism, charac-
result in volume depletion and acute kidney injury. Neuro- terized by high PTH levels and subsequent release of cal-
logic manifestations range from mild drowsiness to weak- cium from bone. Therapy may include intravenous vitamin
ness, depression, lethargy, stupor, and coma in severe D, which further increases the serum calcium concentra-
hypercalcemia. Ventricular ectopy and idioventricular tion. Another type of hypercalcemia occurs when PTH
rhythm occur and can be accentuated by digitalis. levels are low. Bone turnover is decreased, which results in
a low buffering capacity for calcium. When calcium is
B. Laboratory Findings administered in calcium-containing phosphate binders or
dialysate, or when vitamin D is administered, hypercalce-
The ionized calcium exceeds 1.32 mmol/L. A high serum
mia results. Hypercalcemia in dialysis patients usually
chloride concentration and a low serum phosphate con-
occurs in the presence of hyperphosphatemia, and meta-
centration in a ratio > 33:1 (or > 102 if SI units are utilized)
static calcification may occur. Malignancy should be con-
suggests primary hyperparathyroidism where PTH
sidered as a cause of the hypercalcemia.
decreases proximal tubular phosphate reabsorption. A low
serum chloride concentration with a high serum bicarbon-
ate concentration, along with elevated BUN and creatinine, ▶▶When to Refer
suggests milk-alkali syndrome. Severe hypercalcemia (> 15 • Patients may require referral to an oncologist or endo-
mg/dL [or > 3.75 mmol/L]) generally occurs in malig- crinologist depending on the underlying cause of
nancy. More than 300 mg (or > 7.5 mmol) per day of uri- hypercalcemia.
nary calcium excretion suggests hypercalciuria; < 100 mg • Patients with granulomatous diseases (eg, tuberculosis
(or < 2.5 mmol) per day suggests hypocalciuria. Hypercal- and other chronic infections, granulomatosis with poly-
ciuric patients—such as those with malignancy or those angiitis [formerly Wegener granulomatosis], sarcoid-
receiving oral active vitamin D therapy—may easily osis) may require assistance from infectious disease
develop hypercalcemia in case of volume depletion. Serum specialists, rheumatologists, or pulmonologists.
phosphate may or may not be low, depending on the cause.
Hypocalciuric hypercalcemia occurs in milk-alkali syn-
drome, thiazide diuretic use, and familial hypocalciuric ▶▶When to Admit
hypercalcemia. • Patients with symptomatic or severe hypercalcemia
The chest radiograph may reveal malignancy or granu- require immediate treatment.
lomatous disease. The ECG shows a shortened QT interval.
• Unexplained hypercalcemia with associated conditions,
Measurements of PTH and PTHrP help distinguish
such as acute kidney injury or suspected malignancy,
between hyperparathyroidism (elevated PTH) and malig-
may require urgent treatment and expedited evaluation.
nancy-associated hypercalcemia (suppressed PTH, ele-
vated PTHrP).
Bech A et al. Denosumab for tumor-induced hypercalcemia
▶▶Treatment complicated by renal failure. Ann Intern Med. 2012 Jun
19;156(12):906–7. [PMID: 22711097]
Until the primary cause can be identified and treated, renal Crowley R et al. How to approach hypercalcaemia. Clin Med.
excretion of calcium is promoted through aggressive 2013 Jun;13(3):287–90. [PMID: 23760705]
hydration and forced calciuresis. The tendency in hyper- Lindner G et al. Hypercalcemia in the ED: prevalence, etiology,
calcemia is hypovolemia from nephrogenic DI. In dehy- and outcome. Am J Emerg Med. 2013 Apr;31(4):657–60.
[PMID: 23246111]
drated patients with normal cardiac and kidney function,

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 875 7/11/14 5:23 PM
876 CMDT 2015 Chapter 21

Marcocci C et al. Clinical practice. Primary hyperparathyroid- Table 21–9.  Causes of hypophosphatemia.
ism. N Engl J Med. 2011 Dec 22;365(25):2389–97. [PMID:
22187986] Diminished supply or absorption
Rosner MH et al. Onco-nephrology: the pathophysiology and  Starvation
treatment of malignancy-associated hypercalcemia. Clin J   Parenteral alimentation with inadequate phosphate content
Am Soc Nephrol. 2012 Oct;7(10):1722–9. [PMID: 22879438]   Malabsorption syndrome, small bowel bypass
  Absorption blocked by oral antacids with aluminum or
 magnesium
▼▼   Vitamin D–deficient and vitamin D–resistant osteomalacia
DISORDERS OF PHOSPHORUS Increased loss
CONCENTRATION   Phosphaturic drugs: theophylline, diuretics, bronchodilators,
 corticosteroids
Plasma phosphorus is mainly inorganic phosphate and   Hyperparathyroidism (primary or secondary)
represents a small fraction (< 0.2%) of total body phosphate.  Hyperthyroidism
Important determinants of plasma inorganic phosphate   Renal tubular defects with excessive phosphaturia (congenital,
are renal excretion, intestinal absorption, and shift between   induced by monoclonal gammopathy, heavy metal
the intracellular and extracellular spaces. The kidney is the   poisoning), alcoholism
most important regulator of the serum phosphate level.   Hypokalemic nephropathy
PTH decreases reabsorption of phosphate in the proximal   Inadequately controlled diabetes mellitus
tubule while 1,25-dihydroxyvitamin D3 increases reabsorp-   Hypophosphatemic rickets
  Phosphatonins of oncogenic osteomalacia (eg, FGF23
tion. Renal proximal tubular reabsorption of phosphate is
 production)
decreased by volume expansion, corticosteroids, and prox- Intracellular shift of phosphorus
imal tubular dysfunction (as in Fanconi syndrome). Fibro-   Administration of glucose
blast growth factor 23 (FGF23) is a potent phosphaturic   Anabolic steroids, estrogen, oral contraceptives, beta-adrenergic
hormone. Intestinal absorption of phosphate is facilitated   agonists, xanthine derivatives
by active vitamin D. PTH stimulates phosphate release   Hungry bone syndrome
from bone and renal phosphate excretion; primary hyper-   Respiratory alkalosis
parathyroidism can lead to hypophosphatemia and deple-   Salicylate poisoning
tion of bone phosphate stores. By contrast, growth hormone Electrolyte abnormalities
 Hypercalcemia
augments proximal tubular reabsorption of phosphate.
 Hypomagnesemia
Cellular phosphate uptake is stimulated by various factors   Metabolic alkalosis
and conditions, including alkalemia, insulin, epinephrine, Abnormal losses followed by inadequate repletion
feeding, hungry bone syndrome, and accelerated cell   Diabetes mellitus with acidosis, particularly during aggressive
proliferation.  therapy
Phosphorus metabolism and homeostasis are inti-   Recovery from starvation or prolonged catabolic state
mately related to calcium metabolism. See sections on   Chronic alcoholism, particularly during restoration of nutrition;
metabolic bone disease in Chapter 26.   associated with hypomagnesemia
  Recovery from severe burns

FGF23, fibroblast growth factor 23.


HYPOPHOSPHATEMIA

ESSENTIALS OF DIAGNOSIS Serum phosphate levels decrease transiently after food


intake, thus fasting samples are recommended for accu-
▶▶ Severe hypophosphatemia may cause tissue racy. Moderate hypophosphatemia (1.0–2.4 mg/dL [or
hypoxia and rhabdomyolysis. 0.32–0.79 mmol/L]) occurs commonly in hospitalized
▶▶ Renal loss of phosphate can be diagnosed by patients and may not reflect decreased phosphate stores.
measuring urinary phosphate excretion and by In severe hypophosphatemia (< 1 mg/dL [or < 0.32
calculating maximal tubular phosphate reabsorp- mmol/L]), the affinity of hemoglobin for oxygen increases
tion rate (TmP/GFR). through a decrease in the erythrocyte 2,3-biphosphoglyc-
erate concentration, impairing tissue oxygenation and cell
▶▶ PTH and FGF23 are the major factors that decrease metabolism and resulting in muscle weakness or even
TmP/GFR, leading to renal loss of phosphate. rhabdomyolysis. Severe hypophosphatemia is common
and multifactorial in alcoholic patients. In acute alcohol
withdrawal, increased plasma insulin and epinephrine
▶▶General Considerations
along with respiratory alkalosis promote intracellular shift
The leading causes of hypophosphatemia are listed in of phosphate. Vomiting, diarrhea, and poor dietary intake
Table 21–9. Hypophosphatemia may occur in the presence contribute to hypophosphatemia. Chronic alcohol use
of normal phosphate stores. Serious depletion of body results in a decrease in the renal threshold of phosphate
phosphate stores may exist with low, normal, or high excretion. This renal tubular dysfunction reverses after a
serum phosphate concentrations. month of abstinence. Patients with chronic obstructive

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 876 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 877

pulmonary disease and asthma commonly have hypophos- symptomatic hypophosphatemia (< 1 mg/dL [or < 0.32
phatemia, attributed to xanthine derivatives causing shifts mmol/L]), an infusion should provide 279–310 mg/12 h
of phosphate intracellularly and the phosphaturic effects of (or 9–10 mmol/12 h) until the serum phosphorus exceeds
beta-adrenergic agonists, loop diuretics, xanthine deriva- 1 mg/dL and the patient can be switched to oral therapy.
tives, and corticosteroids. Refeeding or glucose administra- The infusion rate should be decreased if hypotension
tion to phosphate-depleted patients may cause fatal occurs. Monitoring of plasma phosphate, calcium, and
hypophosphatemia. potassium every 6 hours is necessary because the response
to phosphate supplementation is not predictable. Magne-
sium deficiency often coexists and should be treated.
▶▶Clinical Findings Contraindications to phosphate replacement include
hypoparathyroidism, advanced CKD, tissue damage and
A. Symptoms and Signs necrosis, and hypercalcemia. When an associated hyper-
Acute, severe hypophosphatemia (< 1.0 mg/dL [or < 0.32 glycemia is treated, phosphate accompanies glucose into
mmol/L]) can lead to rhabdomyolysis, paresthesias, and cells, and hypophosphatemia may ensue.
encephalopathy (irritability, confusion, dysarthria, sei-
zures, and coma). Respiratory failure or failure to wean ▶▶When to Refer
from mechanical ventilation may occur as a result of dia-
phragmatic weakness. Arrhythmias and heart failure are • Patients with refractory hypophosphatemia with
uncommon but serious manifestations. Hematologic mani- increased urinary phosphate excretion may require
festations include acute hemolytic anemia from erythrocyte evaluation by an endocrinologist (for such conditions
fragility, platelet dysfunction with petechial hemorrhages, as hyperparathyroidism and vitamin D disorders) or a
and impaired chemotaxis of leukocytes (leading to nephrologist (for such conditions as renal tubular
increased susceptibility to gram-negative sepsis). defects).
Chronic severe depletion may cause anorexia, pain in • Patients with decreased gastrointestinal absorption may
muscles and bones, and fractures. require referral to a gastroenterologist.

B. Laboratory Findings ▶▶When to Admit


Urine phosphate excretion is a useful clue in the evaluation
Patients with severe or refractory hypophosphatemia will
of hypophosphatemia. The normal renal response to hypo-
require intravenous phosphate.
phosphatemia is decreased urinary phosphate excretion to
< 100 mg/d. The fractional excretion of phosphate (FEPO4)
should be < 5%. The main factors regulating FEPO4 are Bacchetta J et al. Evaluation of hypophosphatemia: lessons from
patients with genetic disorders. Am J Kidney Dis. 2012
PTH and phosphate intake. Increased PTH or phosphate Jan;59(1):152–9. [PMID: 22075221]
intake decreases FEPO4 (ie, more phosphate is excreted into Carpenter TO. The expanding family of hypophosphatemic
the urine). syndromes. J Bone Miner Metab. 2012 Jan;30(1):1–9. [PMID:
Measurement of plasma PTH or PTHrP levels may be 22167381]
helpful. The clinical utility of serum FGF levels is undeter- Felsenfeld AJ et al. Approach to treatment of hypophosphatemia.
mined except in uncommon diseases. Am J Kidney Dis. 2012 Oct;60(4):655–61. [PMID: 22863286]
Imel EA et al. Approach to the hypophosphatemic patient. J Clin
Other clinical features may be suggestive of hypophos- Endocrinol Metab. 2012 Mar;97(3):696–706. [PMID:
phatemia, such as hemolytic anemia and rhabdomyolysis. 22392950]
Fanconi syndrome may present with any combination of Suzuki S et al. Hypophosphatemia in critically ill patients. J Crit
uricosuria, aminoaciduria, normoglycemic glucosuria, Care. 2013 Aug;28(4):536.e9–19. [PMID: 23265292]
normal anion gap metabolic acidosis, and phosphaturia. In
chronic hypophosphatemia, radiographs and bone biopsies
show changes resembling osteomalacia. HYPERPHOSPHATEMIA

▶▶Treatment ESSENTIALS OF DIAGNOSIS


Hypophosphatemia can be prevented by including phos-
phate in repletion and maintenance fluids. A rapid decline ▶▶ Advanced CKD is the most common cause.
in calcium levels can occur with parenteral administration
of phosphate; oral replacement of phosphate is preferable.
▶▶ Hyperphosphatemia in the presence of hypercal-
Moderate hypophosphatemia (1.0–2.5 mg/dL [or 0.32– cemia imposes a high risk of metastatic
0.79 mmol/L]) is usually asymptomatic and does not calcification.
require treatment. The hypophosphatemia in patients with
diabetic ketoacidosis (DKA) will usually correct with nor-
▶▶General Considerations
mal dietary intake. Chronic hypophosphatemia can be
treated with oral phosphate repletion. Mixtures of sodium Advanced CKD with decreased urinary excretion of phos-
and potassium phosphate salts may be given to provide phate is the most common cause of hyperphosphatemia.
0.5–1 g (16–32 mmol) of phosphate per day. For severe, Other causes are listed in Table 21–10.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 877 7/11/14 5:23 PM
878 CMDT 2015 Chapter 21

Table 21–10.  Causes of hyperphosphatemia. Leaf DE et al. A physiologic-based approach to the evaluation of
a patient with hyperphosphatemia. Am J Kidney Dis. 2013
Massive load of phosphate into the extracellular fluid Feb;61(2):330–6. [PMID: 22938849]
  Exogenous sources Lee R et al. Disorders of phosphorus homeostasis. Curr Opin
  Hypervitaminosis D Endocrinol Diabetes Obes. 2010 Dec;17(6):561–7. [PMID:
   Laxatives or enemas containing phosphate 20962635]
   Intravenous phosphate supplement Orrego JJ et al. Hyperphosphatemia. Endocr Pract. 2010 May–
  Endogenous sources Jun;16(3):524–5. [PMID: 20551010]
   Rhabdomyolysis (especially if chronic kidney disease coexists) Prié D et al. Genetic disorders of renal phosphate transport. N
Engl J Med. 2010 Jun 24;362(25):2399–409. [PMID: 20573928]
   Cell lysis by chemotherapy of malignancy, particularly
  lymphoproliferative diseases
   Metabolic acidosis (lactic acidosis, ketoacidosis) ▼▼
   Respiratory acidosis (phosphate incorporation into cells is DISORDERS OF MAGNESIUM
  disturbed) CONCENTRATION
Decreased excretion into urine
  Chronic kidney disease Normal plasma magnesium concentration is 1.8–3.0 mg/
  Acute kidney injury dL (or 0.75–1.25 mmol/L), with about one-third bound to
 Hypoparathyroidism protein and two-thirds existing as free cation. Magnesium
 Pseudohypoparathyroidism excretion is via the kidney. Magnesium’s physiologic effects
 Acromegaly on the nervous system resemble those of calcium.
Pseudohyperphosphatemia Altered magnesium concentration usually provokes an
  Multiple myeloma associated alteration of Ca2+. Both hypomagnesemia and
 Hyperbilirubinemia hypermagnesemia can decrease PTH secretion or action.
 Hypertriglyceridemia
Severe hypermagnesemia (> 5 mg/dL [or 2.1 mmol/L])
  Hemolysis in vitro
suppresses PTH secretion with consequent hypocalcemia;
this disorder is typically seen only in patients receiving
magnesium therapy for preeclampsia. Severe hypomagne-
▶▶Clinical Findings semia causes PTH resistance in end-organs and eventually
A. Symptoms and Signs decreased PTH secretion in severe cases.

The clinical manifestations are those of the underlying HYPOMAGNESEMIA


disorder or associated condition.

B. Laboratory Findings ESSENTIALS OF DIAGNOSIS


In addition to elevated phosphate, blood chemistry abnor-
malities are those of the underlying disease. ▶▶ Serum concentration of magnesium may not be
decreased even in the presence of magnesium
▶▶Treatment depletion. Check urinary magnesium excretion if
Treatment is directed at the underlying cause. Exogenous renal magnesium wasting is suspected.
sources of phosphate, including enteral or parenteral nutri- ▶▶ Causes neurologic symptoms and arrhythmias.
tion and medications, should be reduced or eliminated. ▶▶ Impairs release of PTH.
Dietary phosphate absorption can be reduced by oral phos-
phate binders, such as calcium carbonate, calcium acetate,
sevelamer carbonate, lanthanum carbonate, and aluminum ▶▶General Considerations
hydroxide. Sevelamer, lanthanum, and aluminum may be
used in patients with hypercalcemia, although aluminum Causes of hypomagnesemia are listed in Table 21–11. Nor-
use should be limited to a few days because of the risk of momagnesemia does not exclude magnesium depletion
aluminum accumulation and neurotoxicity. In acute kidney because only 1% of total body magnesium is in the extra-
injury and advanced CKD, dialysis will reduce serum cellular fluid (ECF). Hypomagnesemia and hypokalemia
phosphate. share many etiologies, including diuretics, diarrhea, alco-
holism, aminoglycosides, and amphotericin. Renal potas-
▶▶When to Admit sium wasting also occurs from hypomagnesemia, and is
refractory to potassium replacement until magnesium is
Patients with acute severe hyperphosphatemia require hos- repleted. Hypomagnesemia also suppresses PTH release
pitalization for emergent therapy, possibly including dialy- and causes end-organ resistance to PTH and low 1,25-dihy-
sis. Concomitant illnesses, such as acute kidney injury or droxyvitamin D3 levels. The resultant hypocalcemia is
cell lysis, may necessitate admission. refractory to calcium replacement until the magnesium is
normalized. Molecular mechanisms of magnesium wasting
Howard SC et al. The tumor lysis syndrome. N Engl J Med. 2011 have been revealed in some hereditary disorders. The FDA
May 12;364(19):1844–54. [PMID: 21561350] has issued a warning about hypomagnesemia for patients

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 878 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 879

hypomagnesemia can be treated with 1–2 g of magnesium


Table 21–11.  Causes of hypomagnesemia. sulfate in 10 mL of dextrose 5% solution pushed intrave-
Diminished absorption or intake nously over 15 minutes. Severe, non–life-threatening defi-
  Malabsorption, chronic diarrhea, laxative abuse ciency can be treated at a rate to 1–2 g/h over 3–6 hours.
  Proton pump inhibitors Magnesium sulfate may also be given intramuscularly in a
  Prolonged gastrointestinal suction dosage of 200–800 mg/d (8–33 mmol/d) in four divided
  Small bowel bypass doses. Serum levels must be monitored daily and dosage
 Malnutrition adjusted to keep the concentration from rising above 3 mg/
 Alcoholism dL (1.23 mmol/L). Tendon reflexes may be checked for
  Total parenteral alimentation with inadequate Mg2+ content hyporeflexia of hypermagnesemia. K+ and Ca2+ replace-
Increased renal loss
ment may be required, but patients with hypokalemia and
  Diuretic therapy (loop diuretics, thiazide diuretics)
  Hyperaldosteronism, Gitelman syndrome (a variant of Bartter hypocalcemia of hypomagnesemia do not recover without
 syndrome) magnesium supplementation.
  Hyperparathyroidism, hyperthyroidism Patients with normal kidney function can excrete
 Hypercalcemia excess magnesium; hypermagnesemia should not develop
  Volume expansion with replacement dosages. In patients with CKD, magne-
  Tubulointerstitial diseases sium replacement should be done cautiously to avoid
  Transplant kidney hypermagnesemia. Reduced doses (50–75% dose reduc-
  Drugs (aminoglycoside, cetuximab, cisplatin, amphotericin tion) and more frequent monitoring (at least twice daily)
  B, pentamidine)
are indicated.
Others
  Diabetes mellitus
  Post-parathyroidectomy (hungry bone syndrome)
  Respiratory alkalosis Ayuk J et al. How should hypomagnesaemia be investigated and
 Pregnancy treated? Clin Endocrinol (Oxf). 2011 Dec;75(6):743–6.
[PMID: 21569071]
Blasco LM et al. Chronic cyclic nonnephrogenic magnesium
depletion without losses. N Engl J Med. 2012 May
10;366(19):1845–6. [PMID: 22571217]
taking proton pump inhibitors. The presumed mechanism Danziger J et al. Proton-pump inhibitor use is associated with
is decreased intestinal magnesium absorption, but it is not low serum magnesium concentrations. Kidney Int. 2013
clear why this complication develops in only a small frac- Apr;83(4):692–9. [PMID: 23325090]
Dimke H et al. Evaluation of hypomagnesemia: lessons from
tion of patients taking these medications.
disorders of tubular transport. Am J Kidney Dis. 2013
Aug;62(2):377–83. [PMID: 23201160]
▶▶Clinical Findings
A. Symptoms and Signs HYPERMAGNESEMIA
Common symptoms are those of hypokalemia and hypo-
calcemia, with weakness and muscle cramps. Marked neu-
romuscular and central nervous system hyperirritability ESSENTIALS OF DIAGNOSIS
may produce tremors, athetoid movements, jerking, nys-
tagmus, Babinski response, confusion, and disorientation. ▶▶ Often associated with advanced CKD and chronic
Cardiovascular manifestations include hypertension, intake of magnesium-containing drugs.
tachycardia, and ventricular arrhythmias.

B. Laboratory Findings ▶▶General Considerations


Urinary excretion of magnesium exceeding 10–30 mg/d or Hypermagnesemia is almost always the result of advanced
a fractional excretion > 2% indicates renal magnesium CKD and impaired magnesium excretion. Antacids and
wasting. Hypocalcemia and hypokalemia are often present. laxatives are underrecognized sources of magnesium. Preg-
The ECG shows a prolonged QT interval, due to lengthen- nant patients may have severe hypermagnesemia from
ing of the ST segment. PTH secretion is often suppressed intravenous magnesium for preeclampsia and eclampsia.
(see Hypocalcemia). Magnesium replacement should be done cautiously in
patients with CKD; dose reductions up to 75% may be
necessary to avoid hypermagnesemia.
▶▶Treatment
Magnesium oxide, 250–500 mg orally once or twice daily, is ▶▶Clinical Findings
useful for treating chronic hypomagnesemia. Symptomatic
hypomagnesemia requires intravenous magnesium sulfate A. Symptoms and Signs
1–2 g over 5–60 minutes mixed in either dextrose 5% or Muscle weakness, decreased deep tendon reflexes, mental
0.9% normal saline. Torsades de pointes in the setting of obtundation, and confusion are characteristic manifestations.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 879 7/11/14 5:23 PM
880 CMDT 2015 Chapter 21

Weakness, flaccid paralysis, ileus, urinary retention, and


TYPES OF ACID–BASE DISORDERS
hypotension are noted. Serious findings include respiratory
muscle paralysis and cardiac arrest. There are two types of acid–base disorders: acidosis and
alkalosis. These disorders can be either metabolic (decreased
B. Laboratory Findings or increased HCO3−) or respiratory (decreased or increased
Pco2). Primary respiratory disorders affect blood acidity by
Serum Mg2+ is elevated. In the common setting of CKD,
changes in PCO2, and primary metabolic disorders are dis-
BUN, creatinine, potassium, phosphate, and uric acid may
turbances in HCO3− concentration. A primary disturbance
all be elevated. Serum Ca 2+ is often low. The ECG
is usually accompanied by a compensatory response, but
shows increased PR interval, broadened QRS complexes,
the compensation does not fully correct the pH disturbance
and peaked T waves, probably related to associated
of the primary disorder. If the pH is < 7.40, the primary
hyperkalemia.
process is acidosis, either respiratory (Pco2 > 40 mm Hg) or
metabolic (HCO3− < 24 mEq/L). If the pH is higher than
▶▶Treatment
7.40, the primary process is alkalosis, either respiratory
Exogenous sources of magnesium should be discontinued. (Pco2 < 40 mm Hg) or metabolic (HCO3− > 24 mEq/L). One
Calcium antagonizes Mg2+ and may be given intravenously respiratory or metabolic disorder with its appropriate com-
as calcium chloride, 500 mg or more at a rate of 100 mg (4.1 pensatory response is a simple acid-base disorder.
mmol) per minute. Hemodialysis or peritoneal dialysis
may be necessary to remove magnesium, particularly with MIXED ACID–BASE DISORDERS
severe kidney disease.
Long-term use of magnesium hydroxide and magne- Two or three simultaneous disorders can be present in a
sium sulfate should be avoided in patients with advanced mixed acid-base disorder, but there can never be two pri-
stages of CKD. mary respiratory disorders. Uncovering a mixed acid-base
disorder is clinically important, but requires a methodical
Moe SM. Disorders involving calcium, phosphorus, and magne- approach to acid-base analysis (see box, Step-by-Step Analy-
sium. Prim Care. 2008 Jun;35(2):215–37. [PMID: 18486714] sis of Acid-Base Status). Once the primary disturbance has
Volpe SL. Magnesium in disease prevention and overall health. been determined, the clinician should assess whether the
Adv Nutr. 2013 May 1;4(3):378S–83S. [PMID: 23674807] compensatory response is appropriate (Table 21–12). An
inadequate or an exaggerated response indicates the pres-
▼▼
ence of another primary acid-base disturbance.
ACID–BASE DISORDERS The anion gap should always be calculated for two rea-
sons. First, it is possible to have an abnormal anion gap
Assessment of a patient’s acid–base status requires mea-
even if the sodium, chloride, and bicarbonate levels are
surement of arterial pH, Pco2, and plasma bicarbonate
normal. Second, a large anion gap (> 20 mEq/L) suggests a
(HCO3−). Blood gas analyzers directly measure pH and
primary metabolic acid-base disturbance regardless of the
Pco2. The HCO3− value is calculated from the Henderson–
pH or serum bicarbonate level because a markedly abnor-
Hasselbalch equation:
mal anion gap is never a compensatory response to a respi-
HCO3− ratory disorder. In patients with an increased anion gap
pH = 6.1 +  log  metabolic acidosis, clinicians should calculate the cor-
0.03 ×  PCO2 rected bicarbonate. In increased anion gap acidoses, there
should be a mole for mole decrease in HCO3− as the anion
The total venous CO2 measurement is a more direct deter- gap increases. A corrected HCO3− value higher or lower
mination of HCO3−. Because of the dissociation character- than normal (24 mEq/L) indicates the concomitant pres-
istics of carbonic acid (H2CO3) at body pH, dissolved CO2 ence of metabolic alkalosis or normal anion gap metabolic
is almost exclusively in the form of HCO3−, and for clinical acidosis, respectively.
purposes the total carbon dioxide content is equivalent (±
3 mEq/L) to the HCO3− concentration:
STEP-BY-STEP ANALYSIS OF ACID-BASE
H+ + HCO3− ↔ H2 CO3 ↔ CO2 +  H2 O STATUS
Venous blood gases can provide useful information for Step 1: Determine the primary (or main) disorder—whether it
acid–base assessment since the arteriovenous differences is metabolic or respiratory—from blood pH, HCO3–,
in pH and Pco2 are small and relatively constant. Venous and Pco2 values.
blood pH is usually 0.03–0.04 units lower than arterial Step 2: Determine the presence of mixed acid-base disorders
blood pH, and venous blood Pco2 is 7 or 8 mm Hg higher by calculating the range of compensatory responses
than arterial blood Pco2. Calculated HCO3− concentration (Table 21–12).
Step 3: Calculate the anion gap (Table 21–13).
in venous blood is at most 2 mEq/L higher than arterial
Step 4: Calculate the corrected HCO3– concentration if the
blood HCO3−. Arterial and venous blood gases will not be anion gap is increased (see above).
equivalent during a cardiopulmonary arrest; arterial sam- Step 5: Examine the patient to determine whether the clinical
ples should be obtained for the most accurate measure- signs are compatible with the acid-base analysis.
ments of pH and Pco2.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 880 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 881

Table 21–12.  Primary acid-base disorders and expected compensation.

Disorder Primary Defect Compensatory Response Magnitude of Compensation


Respiratory acidosis
 Acute ↑Pco2 ↑HCO3– ↑ HCO3– 1 mEq/L per 10 mm Hg ↑ Pco2
 Chronic ↑Pco2 ↑HCO3– ↑ HCO3– 3.5 mEq/L per 10 mm Hg ↑ Pco2
Respiratory alkalosis
 Acute ↓Pco2 ↓HCO3– ↓HCO3– 2 mEq/L per 10 mm Hg ↓Pco2
 Chronic ↓Pco2 ↓HCO3– ↓HCO3– 5 mEq/L per 10 mm Hg ↓Pco2
Metabolic acidosis ↓HCO3 –
↓Pco2 ↓Pco2 1.3 mm Hg per 1 mEq/L ↓HCO3–
Metabolic alkalosis ↑HCO3– ↓Pco2 ↑Pco2 0.7 mm Hg per 1 mEq/L ↑HCO3–

Major unmeasured cations are calcium (2 mEq/L), magne-


Adrogué HJ et al. Assessing acid-base disorders. Kidney Int.
2009 Dec;76(12):1239–47. [PMID: 19812535]
sium (2 mEq/L), gamma-globulins, and potassium (4 mEq/L).
Adrogué HJ et al. Secondary responses to altered acid-base sta- Major unmeasured anions are albumin (2 mEq/L per g/dL),
tus: the rules of engagement. J Am Soc Nephrol. 2010 phosphate (2 mEq/L), sulfate (1 mEq/L), lactate (1–2 mEq/L),
Jun;21(6):920–3. [PMID: 20431042] and other organic anions (3–4 mEq/L). Traditionally, the
Berend K. Acid-base pathophysiology after 130 years: confusing,
irrational and controversial. J Nephrol. 2013 Mar–
Apr;26(2):254–65. [PMID: 22976522]
Dzierba AL et al. A practical approach to understanding acid-
base abnormalities in critical illness. J Pharm Pract. 2011
Table 21–13.  Anion gap in metabolic acidosis.1
Feb;24(1):17–26. Erratum in: J Pharm Pract. 2011 Oct;24(5):515.
[PMID: 21507871] Decreased (< 6 mEq)
Wiener SW. Toxicologic acid-base disorders. Emerg Med Clin   Hypoalbuminemia (decreased unmeasured anion)
North Am. 2014 Feb;32(1):149–65. [PMID: 24275173]
  Plasma cell dyscrasias
   Monoclonal protein (cationic paraprotein) (accompanied by
   chloride and bicarbonate)
METABOLIC ACIDOSIS   Bromide intoxication
Increased (> 12 mEq)
  Metabolic anion
ESSENTIALS OF DIAGNOSIS   Diabetic ketoacidosis
  Alcoholic ketoacidosis
  Lactic acidosis
▶▶ Decreased HCO3– with acidemia.    Chronic kidney disease (advanced stages) (PO43–, SO42–)
▶▶ Classified into increased anion gap acidosis and   Starvation
normal anion gap acidosis.    Metabolic alkalosis (increased number of negative charges
  on protein)
▶▶ Lactic acidosis, ketoacidosis, and toxins produce    5-oxoproline acidosis from acetaminophen toxicity
metabolic acidoses with the largest anion gaps.   Drug or chemical anion
▶▶ Normal anion gap acidosis is mainly caused by   Salicylate intoxication
   Sodium carbenicillin therapy
gastrointestinal HCO3– loss or RTA. Urinary anion
   Methanol (formic acid)
gap may help distinguish between these causes.    Ethylene glycol (oxalic acid)
Normal (6–12 mEq)
  Loss of HCO3
▶▶General Considerations   Diarrhea
The hallmark of metabolic acidosis is decreased HCO3–.    Recovery from diabetic ketoacidosis
   Pancreatic fluid loss, ileostomy (unadapted)
Metabolic acidoses are classified by the anion gap, usually
   Carbonic anhydrase inhibitors
normal or increased (Table 21–13). The anion gap is the   Chloride retention
difference between readily measured anions and cations.    Renal tubular acidosis
In plasma,    Ileal loop bladder
  Administration of HCl equivalent or NH4Cl
Unmeasured Unmeasured    Arginine and lysine in parenteral nutrition
 [Na+ ] + cations = HCO3− + Cl− + anions
1
Reference ranges for anion gap may vary based on differing labo-
Anion gap = Na+ − (HCO3− + Cl− ) ratory methods.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 881 7/11/14 5:23 PM
882 CMDT 2015 Chapter 21

normal anion gap has been 12 ± 4 mEq/L. With current Idiopathic lactic acidosis, usually in debilitated patients,
auto-analyzers, the reference range may be lower (6 ± 1 has an extremely high mortality rate. (For treatment of
mEq/L), primarily from an increase in Cl– values. Despite lactic acidosis, see below and Chapter 27.)
its usefulness, the anion gap can be misleading. Non–acid-
base disorders may cause errors in anion gap interpreta- B. Diabetic Ketoacidosis (DKA)
tion; these disorders including hypoalbuminemia,
DKA is characterized by hyperglycemia and metabolic
hypernatremia, or hyponatremia; antibiotics (eg, carbeni-
acidosis with an increased anion gap:
cillin is an unmeasured anion; polymyxin is an unmea-
sured cation) may also cause errors in anion gap
interpretation. Although not usually associated with meta- H+ + B− + NaHCO3 ↔ CO2 +  NaB +  H2 O
bolic acidosis, a decreased anion gap can occur because of
a reduction in unmeasured anions or an increase in where B− is beta-hydroxybutyrate or acetoacetate, the
unmeasured cations. In hypoalbuminemia, a 2 mEq/L ketones responsible for the increased anion gap. The anion
decrease in anion gap will occur for every 1 g/dL decline in gap should be calculated from the measured serum electro-
serum albumin. lytes; correction of the serum sodium for the dilutional
effect of hyperglycemia will exaggerate the anion gap. Dia-
INCREASED ANION GAP ACIDOSIS betics with ketoacidosis may have lactic acidosis from tis-
(Increased Unmeasured Anions) sue hypoperfusion and increased anaerobic metabolism.
During the recovery phase of DKA, a hyperchloremic
Normochloremic metabolic acidosis generally results from non-anion gap acidosis can develop because saline resusci-
addition of organic acids such as lactate, acetoacetate, beta- tation results in chloride retention, restoration of GFR, and
hydroxybutyrate, and exogenous toxins. Other anions such ketoaciduria. Ketone salts (NaB) are formed as bicarbonate
as isocitrate, alpha-ketoglutarate, malate and d-lactate, may is consumed:
contribute to the anion gap of lactic acidosis, DKA, and
acidosis of unknown etiology. Uremia causes an increased HB + NaHCO3 → NaB + H2CO3
anion gap metabolic acidosis from unexcreted organic
acids and anions. The kidney reabsorbs ketone anions poorly but can
compensate for the loss of anions by increasing the reab-
A. Lactic Acidosis sorption of Cl−.
Patients with DKA and normal kidney function may
Lactic acid is formed from pyruvate in anaerobic glycolysis, have marked ketonuria and severe metabolic acidosis but
typically in tissues with high rates of glycolysis, such as gut only a mildly increased anion gap. Thus, the size of the
(responsible for over 50% of lactate production), skeletal anion gap correlates poorly with the severity of the DKA;
muscle, brain, skin, and erythrocytes. Normally, lactate the urinary loss of Na+ or K+ salts of beta-hydroxybutyrate
levels remain low (1 mEq/L) because of metabolism of will lower the anion gap without altering the H+ excretion
lactate principally by the liver through gluconeogenesis or or the severity of the acidosis. Urine dipsticks for ketones
oxidation via the Krebs cycle. The kidneys metabolize test primarily for acetoacetate and, to a lesser degree, ace-
about 30% of lactate. tone but not the predominant ketoacid, beta-hydroxybu-
In lactic acidosis, lactate levels are at least 4–5 mEq/L tyrate. Dipstick tests for ketones may become more positive
but commonly 10–30 mEq/L. There are two basic types of even as the patient improves due to the metabolism of
lactic acidosis. beta-hydroxybutyrate. Thus, the patient’s clinical status
Type A (hypoxic) lactic acidosis is more common, and pH are better markers of improvement than the anion
resulting from decreased tissue perfusion; cardiogenic, gap or ketone levels.
septic, or hemorrhagic shock; and carbon monoxide or
cyanide poisoning. These conditions increase peripheral
C. Alcoholic Ketoacidosis
lactic acid production and decrease hepatic metabolism of
lactate as liver perfusion declines. Chronically malnourished patients who consume large
Type B lactic acidosis may be due to metabolic causes quantities of alcohol daily may develop alcoholic ketoaci-
(eg, diabetes, ketoacidosis, liver disease, kidney disease, dosis. Most of these patients have mixed acid–base disor-
infection, leukemia, or lymphoma) or toxins (eg, ethanol, ders (10% have a triple acid–base disorder). Although
methanol, salicylates, isoniazid, or metformin). Propylene decreased HCO3− is usual, 50% of the patients may have
glycol can cause lactic acidosis from decreased liver metab- normal or alkalemic pH. Three types of metabolic acidosis
olism; it is used as a vehicle for intravenous drugs, such as are seen in alcoholic ketoacidosis: (1) Ketoacidosis is due
nitroglycerin, etomidate, and diazepam. Parenteral nutri- to beta-hydroxybutyrate and acetoacetate excess. (2) Lac-
tion without thiamine causes severe refractory lactic acido- tic acidosis: Alcohol metabolism increases the NADH:NAD
sis from deranged pyruvate metabolism. Patients with ratio, causing increased production and decreased utili-
short bowel syndrome may develop d-lactic acidosis with zation of lactate. Accompanying thiamine deficiency,
encephalopathy due to carbohydrate malabsorption and which inhibits pyruvate carboxylase, further enhances
subsequent fermentation by colonic bacteria. lactic acid production in many cases. Moderate to severe
Nucleoside analog reverse transcriptase inhibitors can elevations of lactate (> 6 mmol/L) are seen with concomi-
cause type B lactic acidosis due to mitochondrial toxicity. tant disorders such as sepsis, pancreatitis, or hypoglycemia.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 882 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 883

(3) Hyperchloremic acidosis from bicarbonate loss in the A. Gastrointestinal HCO3− Loss
urine is associated with ketonuria (see above). Metabolic
alkalosis occurs from volume contraction and vomiting. The gastrointestinal tract secretes bicarbonate at multiple
Respiratory alkalosis results from alcohol withdrawal, pain, sites. Small bowel and pancreatic secretions contain large
or associated disorders such as sepsis or liver disease. Half amounts of HCO3−; massive diarrhea or pancreatic drain-
of the patients have hypoglycemia or hyperglycemia. When age can result in HCO3− loss. Hyperchloremia occurs
serum glucose levels are > 250 mg/dL (>13.88 mmol/L), because the ileum and colon secrete HCO3– in exchange
the distinction from DKA is difficult. The absence of a for Cl− by countertransport. The resultant volume con-
diabetic history and normoglycemia after initial therapy traction causes increased Cl− retention by the kidney in
support the diagnosis of alcoholic ketoacidosis. the setting of decreased HCO3−. Patients with ureterosig-
moidostomies can develop hyperchloremic metabolic aci-
D. Toxins dosis because the colon secretes HCO3− in the urine in
exchange for Cl−.
(See also Chapter 38.) Multiple toxins and drugs increase the
anion gap by increasing endogenous acid production. Com- B. Renal Tubular Acidosis (RTA)
mon examples include methanol (metabolized to formic
acid), ethylene glycol (glycolic and oxalic acid), and salicy- Hyperchloremic acidosis with a normal anion gap and
lates (salicylic acid and lactic acid). The latter can cause a normal (or near normal) GFR, and in the absence of diar-
mixed disorder of metabolic acidosis with respiratory alka- rhea, defines RTA. The defect is either inability to excrete
losis. In toluene poisoning, the metabolite hippurate is rap- H+ (inadequate generation of new HCO3–) or inappropriate
idly excreted by the kidney and may present as a normal reabsorption of HCO3–. Three major types can be differen-
anion gap acidosis. Isopropanol, which is metabolized to tiated by the clinical setting, urinary pH, urinary anion gap
acetone, increases the osmolar gap, but not the anion gap. (see below), and serum K+ level. The pathophysiologic
mechanisms of RTA have been elucidated by identifying
E. Uremic Acidosis the responsible molecules and gene mutations.

As the GFR drops below 15–30 mL/min, the kidneys are 1. Classic distal RTA (type I)—This disorder is characterized
increasingly unable to excrete H+ and organic acids, such as by selective deficiency in H+ secretion in alpha intercalated
phosphate and sulfate, resulting in an increased anion gap cells in the collecting tubule. Despite acidosis, urinary pH
acidosis. Hyperchloremic normal anion gap acidosis devel- cannot be acidified and is above 5.5, which retards the bind-
ops in earlier stages of CKD. ing of H+ to phosphate (H+ + HPO42− → H2PO4) and inhibits
titratable acid excretion. Furthermore, urinary excretion of
NORMAL ANION GAP ACIDOSIS NH4+Cl− is decreased, and the urinary anion gap is positive
(Table 21–14) (see below). Enhanced K+ excretion occurs probably because
there is less competition from H+ in the distal nephron
The two major causes are gastrointestinal HCO3– loss and transport system. Furthermore, hyperaldosteronism occurs
defects in renal acidification (renal tubular acidoses). The in response to renal salt wasting, which will increase potas-
urinary anion gap can differentiate between these causes sium excretion. Nephrocalcinosis and nephrolithiasis are
(see below). often seen in patients with distal RTA since chronic acidosis

Table 21–14.  Hyperchloremic, normal anion gap metabolic acidoses.

Distal H+ Secretion

Urinary NH4+ Urinary


Serum Plus Minimal Titrat- Anion
Renal Defect [K+] Urine pH able Acid Gap Treatment
Gastrointestinal None ↓ < 5.5 ↑↑ Negative + +
Na , K , and HCO3– as required
HCO3– loss
Renal tubular acidosis
  I. Classic distal Distal H+ secretion ↓ > 5.5 ↓ Positive NaHCO3 (1–3 mEq/kg/d)
  II. Proximal secretion Proximal H+ ↓ < 5.5 Normal Positive NaHCO3 or KHCO3 (10–15 mEq/
kg/d), thiazide
  IV. Hyporeninemic Distal Na+ reabsorp- ↑ < 5.5 ↓ Positive Fludrocortisone (0.1–0.5 mg/d),
hypoaldosteronism tion, K+ secretion, dietary K+ restriction, furose-
and H+ secretion mide (40–160 mg/d), NaHCO3
(1–3 mEq/kg/d)

Modified and reproduced, with permission, from Cogan MG. Fluid and Electrolytes: Physiology and Pathophysiology. McGraw-Hill, 1991.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 883 7/11/14 5:23 PM
884 CMDT 2015 Chapter 21

decreases tubular calcium reabsorption. Hypercalciuria, F. Hyperalimentation


alkaline urine, and lowered level of urinary citrate cause
calcium phosphate stones and nephrocalcinosis. Hyperalimentation fluids may contain amino acid solu-
Distal RTA develops as a consequence of paraprotein- tions that acidify when metabolized, such as arginine
emias, autoimmune disease, and drugs and toxins such as hydrochloride and lysine hydrochloride.
amphotericin.
▶▶Assessment of Hyperchloremic Metabolic
2. Proximal RTA (type II)—Proximal RTA is due to a selec- Acidosis by Urinary Anion Gap
tive defect in the proximal tubule’s ability to reabsorb
Increased renal NH4+Cl− excretion to enhance H+ removal
filtered HCO3−. Carbonic anhydrase inhibitors (acetazol-
is the normal physiologic response to metabolic acidosis.
amide) can cause proximal RTA. About 90% of filtered
The daily urinary excretion of NH4Cl can be increased
HCO3− is absorbed by the proximal tubule. A proximal
from 30 mEq to 200 mEq in response to acidosis.
defect in HCO3− reabsorption will overwhelm the distal
The urinary anion gap (Na+ + K+ − Cl−) reflects the abil-
tubule’s limited capacity to reabsorb HCO3−, resulting in
ity of the kidney to excrete NH4Cl. The urinary anion gap
bicarbonaturia and metabolic acidosis. Distal delivery of
differentiates between gastrointestinal and renal causes of
HCO3− declines as the plasma HCO3− level decreases.
hyperchloremic acidosis. If the cause is gastrointestinal
When the plasma HCO3− level drops to 15–18 mEq/L, the
HCO3− loss (diarrhea), renal acidification remains normal
distal nephron can reabsorb the diminished filtered load
and NH4Cl excretion increases, and the urinary anion gap
of HCO3−. Bicarbonaturia resolves, and the urinary pH
is negative. If the cause is distal RTA, the urinary anion gap
can be acidic. Thiazide-induced volume contraction can
is positive, since the basic lesion in the disorder is the inabil-
be used to enhance proximal HCO3− reabsorption, lead-
ity of the kidney to excrete H+ as NH4Cl. In proximal (type
ing to the decrease in distal HCO3− delivery and improve-
II) RTA, the kidney has defective HCO3− reabsorption,
ment of bicarbonaturia and renal acidification. The
leading to increased HCO3− excretion rather than decreased
increased delivery of HCO3− to the distal nephron
NH4Cl excretion; the urinary anion gap is often negative.
increases K+ secretion, and hypokalemia results if a
Urinary pH may not readily differentiate between the
patient is loaded with excess HCO3− and K+ is not ade-
two causes. Despite acidosis, if volume depletion from
quately supplemented. Proximal RTA can exist with other
diarrhea causes inadequate Na+ delivery to the distal neph-
proximal reabsorption defects, such as Fanconi syn-
ron and therefore decreased exchange with H+, urinary pH
drome, resulting in glucosuria, aminoaciduria, phospha-
may not be lower than 5.3. In the presence of this relatively
turia, and uricosuria. Causes include multiple myeloma
high urinary pH, however, H+ excretion continues due to
and nephrotoxic drugs.
buffering of NH3 to NH4+, since the pK of this reaction is
3. Hyporeninemic hypoaldosteronemic RTA (type IV)— as high as 9.1. Potassium depletion, which can accompany
Type IV is the most common RTA in clinical practice. The diarrhea (and surreptitious laxative abuse), may also impair
defect is aldosterone deficiency or antagonism, which renal acidification. Thus, when volume depletion is pres-
impairs distal nephron Na+ reabsorption and K+ and H+ ent, the urinary anion gap is a better measure of ability to
excretion. Renal salt wasting and hyperkalemia are fre- acidify the urine than urinary pH.
quently present. Common causes are diabetic nephropathy, When large amounts of other anions are present in the
tubulointerstitial renal diseases, hypertensive nephroscler- urine, the urinary anion gap may not be reliable. In such a
osis, and AIDS. In patients with these disorders, drugs, situation, NH4+ excretion can be estimated using the uri-
such as ACE inhibitors, spironolactone, and NSAIDs, can nary osmolar gap.
exacerbate the hyperkalemia.
NH4+ excretion (mmol/L) = 0.5 × Urinary osmolar gap
C. Dilutional Acidosis = 0.5 [U osm – 2(U Na+ + U K+) + U urea + U glucose]

Rapid dilution of plasma volume by 0.9% NaCl may cause where urine concentrations and osmolality are in mmol/L.
hyperchloremic acidosis.
▶▶Clinical Findings
D. Recovery from DKA A. Symptoms and Signs
See Increased Anion Gap Acidosis (Increased Unmeasured Symptoms of metabolic acidosis are mainly those of the
Anion). underlying disorder. Compensatory hyperventilation is an
important clinical sign and may be misinterpreted as a
E. Posthypocapnia primary respiratory disorder; Kussmaul breathing (deep,
regular, sighing respirations) may be seen with severe
In prolonged respiratory alkalosis, HCO3− decreases and metabolic acidosis.
Cl− increases from decreased renal NH4+Cl− excretion. If
the respiratory alkalosis is corrected quickly, HCO3− will B. Laboratory Findings
remain low until the kidneys can generate new HCO3−,
which generally takes several days. In the meantime, the Blood pH, serum HCO3–, and Pco2 are decreased. Anion
increased Pco2 with low HCO3− causes metabolic gap may be normal (hyperchloremic) or increased (nor-
acidosis. mochloremic). Hyperkalemia may be seen.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 884 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 885

▶▶Treatment ▶▶When to Admit


A. Increased Anion Gap Acidosis Patients will require emergency department evaluation or
hospital admission depending on the severity of the acido-
Treatment is aimed at the underlying disorder, such as insu-
sis and underlying conditions.
lin and fluid therapy for diabetes and appropriate volume
resuscitation to restore tissue perfusion. The metabolism
of lactate will produce HCO3– and increase pH. Supple- Haque SK et al. Proximal renal tubular acidosis: a not so rare
mental HCO3– is indicated for treatment of hyperkale- disorder of multiple etiologies. Nephrol Dial Transplant. 2012
mia (Table 21–6) and some forms of normal anion gap Dec;27(12):4273–87. [PMID: 23235953]
Kraut JA et al. Approach to the evaluation of a patient with an
acidosis but has been controversial for treatment of
increased serum osmolal gap and high-anion-gap metabolic
increased anion gap metabolic acidosis with respect to effi- acidosis. Am J Kidney Dis. 2011 Sep;58(3):480–4. [PMID:
cacy and safety. Large amounts of HCO3– may have deleteri- 21794966]
ous effects, including hypernatremia, hyperosmolality, Kraut JA et al. Differential diagnosis of nongap metabolic acido-
volume overload, and worsening of intracellular acidosis. sis: value of a systematic approach. Clin J Am Soc Nephrol.
In addition, alkali administration stimulates phospho- 2012 Apr;7(4):671–9. [PMID: 22403272]
Liamis G et al. Pharmacologically-induced metabolic acidosis:
fructokinase activity, thus exacerbating lactic acidosis via
a review. Drug Saf. 2010 May 1;33(5):371–91. [PMID:
enhanced lactate production. Ketogenesis is also aug- 20397738]
mented by alkali therapy. Sia P et al. Type B lactic acidosis associated with multiple
In salicylate intoxication, alkali therapy must be started myeloma. Am J Kidney Dis. 2013 Sep;62(3):633–7. [PMID:
to decrease central nervous system damage unless blood 23759296]
pH is already alkalinized by respiratory alkalosis, since an
increased pH converts salicylate to more impermeable sali-
cylic acid. In alcoholic ketoacidosis, thiamine should be METABOLIC ALKALOSIS
given with glucose to avoid Wernicke encephalopathy. The
bicarbonate deficit can be calculated as follows:
ESSENTIALS OF DIAGNOSIS
HCO3– deficit = 0.5 × body weight in kg × (24 – HCO3–)
Half of the calculated deficit should be administered ▶▶ High HCO3– with alkalemia.
within the first 3–4 hours to avoid overcorrection and vol- ▶▶ Evaluate effective circulating volume by physical
ume overload. In methanol intoxication, inhibition of examination.
alcohol dehydrogenase by fomepizole is now standard care. ▶▶ Check urinary chloride concentration to differenti-
Ethanol had previously been used as a competitive sub- ate saline-responsive alkalosis from saline-unre-
strate for alcohol dehydrogenase, which metabolizes to sponsive alkalosis.
formaldehyde.

B. Normal Anion Gap Acidosis


▶▶Classification
Treatment of RTA is mainly achieved by administration of
Metabolic alkalosis is characterized by high HCO3−.
alkali (either as bicarbonate or citrate) to correct metabolic
Abnormalities that generate HCO3– are called “initiation
abnormalities and prevent nephrocalcinosis and CKD.
factors,” whereas abnormalities that promote renal conser-
Large amounts of oral alkali (10–15 mEq/kg/d)
vation of HCO3– are called “maintenance factors.” Thus,
(Table 21–14) may be required to treat proximal RTA
metabolic alkalosis may remain even after the initiation
because most of the alkali is excreted into the urine, which
factors have resolved.
exacerbates hypokalemia. Thus, a mixture of sodium and
The causes of metabolic alkalosis are classified into two
potassium salts is preferred. Thiazides may reduce the
groups based on “saline responsiveness” using the urine
amount of alkali required, but hypokalemia may develop.
Cl– as a marker for volume status (Table 21–15). Saline-
Treatment of type 1 distal RTA requires less alkali (1–3
responsive metabolic alkalosis is a sign of extracellular
mEq/kg/d) than proximal RTA. Potassium supplementa-
volume contraction, and saline-unresponsive alkalosis
tion may be necessary.
implies excessive total body bicarbonate with either
For type IV RTA, dietary potassium restriction may be
euvolemia or hypervolemia. The compensatory increase in
necessary and potassium-retaining drugs should be with-
Pco2 rarely exceeds 55 mm Hg; higher Pco2 values imply a
drawn. Fludrocortisone may be effective in cases with
superimposed primary respiratory acidosis.
hypoaldosteronism, but should be used with care, prefera-
bly in combination with loop diuretics. In some cases, oral
A. Saline-Responsive Metabolic Alkalosis
alkali supplementation (1–3 mEq/kg/d) may be required.
Much more common than saline-unresponsive alkalosis,
▶▶When to Refer saline-responsive alkalosis is characterized by normoten-
sive extracellular volume contraction and hypokalemia.
Most clinicians will refer patients with renal tubular acidoses Hypotension and orthostasis may be seen. In vomiting or
to a nephrologist for evaluation and possible alkali therapy. nasogastric suction, loss of acid (HCl) initiates the

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 885 7/11/14 5:23 PM
886 CMDT 2015 Chapter 21

Table 21–15.  Metabolic alkalosis.

Saline-Responsive (UCl < 25 mEq/L) Saline-Unresponsive (UCl > 40 mEq/L)


Excessive body bicarbonate content Excessive body bicarbonate content
  Renal alkalosis   Renal alkalosis
  Diuretic therapy   Normotensive
   Poorly reabsorbable anion therapy: carbenicillin,     Bartter syndrome (renal salt wasting and secondary hyperaldosteronism)
   penicillin, sulfate, phosphate    Severe potassium depletion
  Posthypercapnia    Refeeding alkalosis
  Gastrointestinal alkalosis    Hypercalcemia and hypoparathyroidism
   Loss of HCl from vomiting or nasogastric suction  Hypertensive
   Intestinal alkalosis: chloride diarrhea   Endogenous mineralocorticoids
  NaHCO3 (baking soda)    Primary aldosteronism
   Sodium citrate, lactate, gluconate, acetate    Hyperreninism
  Transfusions     Adrenal enzyme (11-beta-hydroxylase and 17-alpha-hydroxylase)
  Antacids    deficiency
Normal body bicarbonate content    Liddle syndrome
   “Contraction alkalosis”   Exogenous alkali
  Exogenous mineralocorticoids
  Licorice

Modified and reproduced, with permission, from Narins RG et al. Diagnostic strategies in disorders of fluid, electrolyte and acid-base homeo-
stasis. Am J Med. 1982 Mar;72(3):496–520.

alkalosis, but volume contraction from Cl− loss maintains B. Saline-Unresponsive Alkalosis
the alkalosis because the kidney avidly reabsorbs Na+ to
restore the ECF. Increased sodium reabsorption necessi- 1. Hyperaldosteronism—Primary hyperaldosteronism
tates increased HCO3− reabsorption proximally, and the causes extracellular volume expansion and hypertension by
urinary pH remains acidic despite alkalemia (paradoxical increasing distal sodium reabsorption. Aldosterone
aciduria). Renal Cl– reabsorption is high, and urine Cl– is increases H+ and K+ excretion, producing metabolic alka-
low (< 10–20 mEq/L). In alkalosis, bicarbonaturia may losis and hypokalemia. In an attempt to decrease extracel-
force Na+ excretion as the accompanying cation even if lular volume, high levels of NaCl are excreted resulting in a
volume depletion is present. Therefore, urine Cl− is pre- high urine Cl− (> 20 mEq/L). Therapy with NaCl will only
ferred to urine Na+ as a measure of extracellular volume. increase volume expansion and hypertension and will not
Diuretics may limit the utility of urine chloride by increas- treat the underlying problem of mineralocorticoid excess.
ing urine chloride and sodium excretion, even in the set- 2. Alkali administration with decreased GFR—The normal
ting of volume contraction. kidney has a substantial capacity for bicarbonate excretion,
Metabolic alkalosis is generally associated with hypoka- protecting against metabolic alkalosis even with large
lemia due to the direct effect of alkalosis on renal potas- HCO3− intake. However, urinary excretion of bicarbonate is
sium excretion and secondary hyperaldosteronism from inadequate in CKD. If large amounts of HCO3− are con-
volume depletion. Hypokalemia exacerbates the metabolic sumed, as with intensive antacid therapy, metabolic alkalo-
alkalosis by increasing bicarbonate reabsorption in the sis will occur. Lactate, citrate, and gluconate can also cause
proximal tubule and hydrogen ion secretion in the distal metabolic alkalosis because they are metabolized to bicar-
tubule. Administration of KCl will correct the disorder. bonate. In milk-alkali syndrome, sustained heavy ingestion
1. Contraction alkalosis—Diuretics decrease extracellular of absorbable antacids and milk causes hypercalcemic kid-
volume from urinary loss of NaCl and water. The plasma ney injury and metabolic alkalosis. Volume contraction
HCO3− concentration increases because the extracellular from renal hypercalcemic effects exacerbates the alkalosis.
fluid volume contracts around a stable total body bicar-
bonate. Contraction alkalosis is the opposite of dilutional ▶▶Clinical Findings
acidosis.
A. Symptoms and Signs
2. Posthypercapnia alkalosis—In chronic respiratory aci-
There are no characteristic symptoms or signs. Orthostatic
dosis, the kidney decreases bicarbonate excretion, increasing
hypotension may be encountered. Concomitant hypokale-
plasma HCO3− concentration (Table 21–12). Hypercapnia
mia may cause weakness and hyporeflexia. Tetany and
directly affects the proximal tubule to decrease NaCl reab-
neuromuscular irritability occur rarely.
sorption, which can cause extracellular volume depletion. If
Pco2 is rapidly corrected, metabolic alkalosis will exist
until the kidney excretes the retained bicarbonate. Many
B. Laboratory Findings
patients with chronic respiratory acidosis receive diuretics, The arterial blood pH and bicarbonate are elevated. With
which further exacerbates the metabolic alkalosis. respiratory compensation, the arterial Pco2 is increased.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 886 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 887

Serum potassium and chloride are decreased. There may acidosis is corrected suddenly, posthypercapnic metabolic
be an increased anion gap. The urine chloride can differen- alkalosis ensues until the kidneys excrete the excess bicar-
tiate between saline-responsive (< 25 mEq/L) and unre- bonate over 2–3 days.
sponsive (> 40 mEq/L) causes.
▶▶Clinical Findings
▶▶Treatment
A. Symptoms and Signs
Mild alkalosis is generally well tolerated. Severe or symp-
tomatic alkalosis (pH > 7.60) requires urgent treatment. With acute onset, somnolence, confusion, mental status
changes, asterixis, and myoclonus may develop. Severe
A. Saline-Responsive Metabolic Alkalosis hypercapnia increases cerebral blood flow, cerebrospinal
fluid pressure, and intracranial pressure; papilledema and
Therapy for saline-responsive metabolic alkalosis is correc- pseudotumor cerebri may be seen.
tion of the extracellular volume deficit with isotonic saline.
Diuretics should be discontinued. H2-blockers or proton B. Laboratory Findings
pump inhibitors may be helpful in patients with alkalosis
from nasogastric suctioning. If pulmonary or cardiovascu- Arterial pH is low and Pco2 is increased. Serum HCO3– is
lar disease prohibits adequate resuscitation, acetazolamide elevated but does not fully correct the pH. If the disorder is
will increase renal bicarbonate excretion. Hypokalemia chronic, hypochloremia is seen.
may develop because bicarbonate excretion may induce
kaliuresis. Severe cases, especially those with reduced kid- ▶▶Treatment
ney function, may require dialysis with low-bicarbonate
dialysate. If opioid overdose is a possible diagnosis or there is no
other obvious cause for hypoventilation, the clinician
B. Saline-Unresponsive Metabolic Alkalosis should consider a diagnostic and therapeutic trial of intra-
venous naloxone (see Chapter 38). In all forms of respira-
Therapy for saline-unresponsive metabolic alkalosis tory acidosis, treatment is directed at the underlying
includes surgical removal of a mineralocorticoid-produc- disorder to improve ventilation.
ing tumor and blockage of aldosterone effect with an ACE
inhibitor or with spironolactone (see Chapter 26). Meta-
Adrogué HJ. Diagnosis and management of severe respiratory
bolic alkalosis in primary aldosteronism can be treated acidosis. Am J Kidney Dis. 2010 Nov;56(5):994–1000. [PMID:
only with potassium repletion. 20673604]
Chebbo A et al. Hypoventilation syndromes. Med Clin North
Feldman M et al. Respiratory compensation to a primary meta- Am. 2011 Nov;95(6):1189–202. [PMID: 22032434]
bolic alkalosis in humans. Clin Nephrol. 2012 Nov;78(5):365–9. Marik PE. The malignant obesity hypoventilation syndrome
[PMID: 22854166] (MOHS). Obes Rev. 2012 Oct;13(10):902–9. [PMID:
Gennari FJ. Pathophysiology of metabolic alkalosis: a new clas- 22708580]
sification based on the centrality of stimulated collecting duct Schwartzstein RM et al. Rising PaCO(2) in the ICU: using a
ion transport. Am J Kidney Dis. 2011 Oct;58(4):626–36. physiologic approach to avoid cognitive biases. Chest. 2011
[PMID: 21849227] Dec;140(6):1638–42. [PMID: 22147823]
Peixoto AJ et al. Treatment of severe metabolic alkalosis in a
patient with congestive heart failure. Am J Kidney Dis. 2013
May;61(5):822–7. [PMID: 23481366]
Yi JH et al. Metabolic alkalosis from unsuspected ingestion: use RESPIRATORY ALKALOSIS (HYPOCAPNIA)
of urine pH and anion gap. Am J Kidney Dis. 2012
Apr;59(4):577–81. [PMID: 22265393] Respiratory alkalosis occurs when hyperventilation
reduces the Pco2, increasing serum pH. The most com-
mon cause of respiratory alkalosis is hyperventilation
RESPIRATORY ACIDOSIS (HYPERCAPNIA) syndrome (Table 21–16), but bacterial septicemia and cir-
rhosis are other common causes. In pregnancy, progester-
Respiratory acidosis results from hypoventilation and sub- one stimulates the respiratory center, producing an average
sequent hypercapnia. Pulmonary and extrapulmonary Pco2 of 30 mm Hg and respiratory alkalosis. Symptoms of
disorders can cause hypoventilation. acute respiratory alkalosis are related to decreased cerebral
Acute respiratory failure is associated with severe aci- blood flow induced by the disorder.
dosis and only a small increase in the plasma bicarbonate. Determination of appropriate metabolic compensation
After 6–12 hours, the primary increase in Pco2 evokes a may reveal an associated metabolic disorder (see Mixed
renal compensation to excrete more acid and to generate Acid–Base Disorders).
more HCO3–; complete metabolic compensation by the As in respiratory acidosis, the metabolic compensation
kidney takes several days. is greater if the respiratory alkalosis is chronic (Table 21–12).
Chronic respiratory acidosis is generally seen in patients Although serum HCO3− is frequently < 15 mEq/L in meta-
with underlying lung disease, such as chronic obstructive bolic acidosis, such a low level in respiratory alkalosis
pulmonary disease. Renal excretion of acid as NH4Cl is unusual and may represent a concomitant primary
results in hypochloremia. When chronic respiratory metabolic acidosis.

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 887 7/11/14 5:23 PM
888 CMDT 2015 Chapter 21

B. Laboratory Findings
Table 21–16.  Causes of respiratory alkalosis.
Arterial blood pH is elevated, and Pco2 is low. Serum
Hypoxia bicarbonate is decreased in chronic respiratory alkalosis.
  Decreased inspired oxygen tension
  High altitude ▶▶Treatment
  Ventilation/perfusion inequality
 Hypotension Treatment is directed toward the underlying cause. In acute
  Severe anemia hyperventilation syndrome from anxiety, the traditional
CNS-mediated disorders treatment of breathing into a paper bag should be discour-
  Voluntary hyperventilation aged because it does not correct Pco2 and may decrease
  Anxiety-hyperventilation syndrome
Po2. Reassurance may be sufficient for the anxious patient,
  Neurologic disease
  Cerebrovascular accident (infarction, hemorrhage)
but sedation may be necessary if the process persists.
 Infection Hyperventilation is usually self-limited since muscle weak-
 Trauma ness caused by the respiratory alkalemia will suppress
 Tumor ventilation. Rapid correction of chronic respiratory alkalo-
  Pharmacologic and hormonal stimulation sis may result in metabolic acidosis as Pco2 is increased in
  Salicylates the setting of a previous compensatory decrease in HCO3−.
  Nicotine
  Xanthines
Curley G et al. Bench-to-bedside review: carbon dioxide. Crit
  Pregnancy (progesterone)
Care. 2010;14(2):220. [PMID: 20497620]
  Hepatic failure Palmer BF. Evaluation and treatment of respiratory alkalosis.
  Gram-negative septicemia Am J Kidney Dis. 2012 Nov;60(5):834–8. [PMID: 22871240]
  Recovery from metabolic acidosis
  Heat exposure
Pulmonary disease
▼▼
  Interstitial lung disease FLUID MANAGEMENT
 Pneumonia
  Pulmonary embolism Daily parenteral maintenance fluids and electrolytes for an
  Pulmonary edema average adult would include at least 2 L of water in the form
Mechanical overventilation of 0.45% saline with 20 mEq/L of potassium chloride.
Adapted, with permission, from Gennari FJ. Respiratory acidosis and
Patients with hypoglycemia, starvation ketosis, or ketoaci-
alkalosis. In: Maxwell and Kleeman’s Clinical Disorders of Fluid and dosis being treated with insulin may require 5% dextrose-
Electrolyte Metabolism, 5th ed. Narins RG (editor). McGraw-Hill, 1994. containing solutions. Guidelines for gastrointestinal fluid
losses are shown in Table 21–17.
Weight loss or gain is the best indication of water bal-
▶▶Clinical Findings ance. Insensible water loss should be considered in febrile
patients. Water loss increases by 100–150 mL/d for each
A. Symptoms and Signs degree of body temperature over 37°C.
In acute cases (hyperventilation), there is light-headedness, In patients requiring maintenance and possibly replace-
anxiety, perioral numbness, and paresthesias. Tetany ment of fluid and electrolytes by parenteral infusion, the
occurs from a low ionized calcium, since severe alkalosis total daily ration should be administered continuously over
increases calcium binding to albumin. 24 hours to ensure optimal utilization.

Table 21–17.  Replacement guidelines for sweat and gastrointestinal fluid losses.

Average Electrolyte Composition Replacement Guidelines per Liter Lost

0.9% 0.45% 7.5% NaHCO3


Na+ K+ Cl– HCO3– Saline Saline KCl (45 mEq
(mEq/L) (mEq/L) (mEq/L) (mEq/L) (mL) (mL) D5W (mL) (mEq/L) HCO3–/amp)
Sweat 30–50 5 50 500 500 5
Gastric secretions 20 10 10 300 700 20
Pancreatic juice 130 5 35 115 400 600 5 2 amps
Bile 145 5 100 25 600 400 5 0.5 amp
Duodenal fluid 60 15 100 10 1000 15 0.25 amp
Ileal fluid 100 10 60 60 600 400 10 1 amp
Colonic diarrhea 1401 10 85 60 1000 10 1 amp

In the absence of diarrhea, colonic fluid Na+ levels are low (40 mEq/L).
1

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 888 7/11/14 5:23 PM
ELECTROLYTE & ACID-BASE DISORDERS CMDT 2015 889

If intravenous fluids are the only source of water, elec- those with critical illness or acute kidney injury, and has
trolytes, and calories for longer than a week, parenteral been associated with worsened outcomes such as prolonged
nutrition containing amino acids, lipids, trace metals, and mechanical ventilation, dependence on dialysis, and long
vitamins may be indicated. (See Chapter 29.) duration of hospitalization with increased mortality.
For parenteral alimentation, 620 mg (20 mmol) of
phosphorus is required for every 1000 nonprotein kcal to Annane D et al. Effects of fluid resuscitation with colloids vs
maintain phosphate balance and to ensure anabolic func- crystalloids on mortality in critically ill patients presenting
tion. For prolonged parenteral fluid maintenance, a daily with hypovolemic shock: the CRISTAL randomized trial.
ration is 620–1240 mg (20–40 mmol) of phosphorus. JAMA. 2013 Nov 6;310(17):1809–17. [PMID: 24108515]
Excessive fluid resuscitation or maintenance is now Myburgh JA et al. Resuscitation fluids. N Engl J Med. 2013 Sep
26;369(13):1243–51. [PMID: 24066745]
viewed as a complication in hospitalized patients, especially

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH21-p0861-p0889.indd 889 7/11/14 5:23 PM
890 CMDT 2015

22
Kidney Disease
Suzanne Watnick, MD
Tonja Dirkx, MD

Kidney disease can be discovered incidentally during a formed elements—crystals, cells, casts, and infecting
routine medical evaluation or with evidence of kidney dys- organisms.
function, such as hypertension, edema, nausea, or hematu- Various findings on the urinalysis are indicative of cer-
ria. The initial approach in both situations should be to tain patterns of kidney disease. A bland (normal) urinary
assess the cause and severity of renal abnormalities. In all sediment is common, especially in CKD and acute disor-
cases this evaluation includes (1) an estimation of disease ders that are not intrinsic to the kidney, such as limited
duration, (2) a careful urinalysis, and (3) an assessment of effective blood flow to the kidney or obstruction of
the glomerular filtration rate (GFR). The history and physi- the urinary outflow tract. Casts are composed of Tamm-
cal examinations, though equally important, are variable Horsfall urinary mucoprotein in the shape of the nephron
among renal syndromes—thus, specific symptoms and segment where they were formed. Heavy proteinuria and
signs are discussed under each disease entity. lipiduria are consistent with the nephrotic syndrome. The
presence of hematuria with dysmorphic red blood cells, red
blood cell casts, and proteinuria is indicative of glomerulo-
▼▼
ASSESSMENT OF KIDNEY DISEASE nephritis. Dysmorphic red blood cells are misshapen dur-
ing abnormal passage from the capillary through the
▶▶Disease Duration glomerular basement membrane (GBM) into the urinary
space of Bowman capsule. Pigmented granular casts and
Kidney disease may be acute or chronic. Acute kidney renal tubular epithelial cells alone or in casts suggest acute
injury is worsening of kidney function over hours to days, tubular necrosis. White blood cells, including neutrophils
resulting in the retention of nitrogenous wastes (such as and eosinophils, white blood cell casts (Table 22–1), red
urea nitrogen) and creatinine in the blood. Retention of blood cells, and small amounts of protein can be found in
these substances is called azotemia. Chronic kidney disease interstitial nephritis and pyelonephritis; Wright and Hansel
(CKD) results from an abnormal loss of kidney function stains can detect eosinophiluria. Pyuria alone can indicate
over months to years. Differentiating between the two is a urinary tract infection. Hematuria and proteinuria are
important for diagnosis, treatment, and outcome. Oliguria discussed more thoroughly below.
is unusual in CKD. Anemia (from low kidney erythropoi-
etin production) is rare in the initial period of acute kidney A. Proteinuria
disease. Small kidneys are most consistent with CKD,
whereas normal to large-size kidneys can be seen with both Proteinuria is defined as excessive protein excretion in the
chronic and acute disease. urine, generally > 150–160 mg/24 h in adults. Significant
proteinuria is a sign of an underlying kidney abnormality,
usually glomerular in origin when > 1–2 g/d. Less than 1 g/d
▶▶Urinalysis
can be due to multiple causes along the nephron segment, as
A urinalysis can provide information similar to a kidney listed below. Proteinuria can be accompanied by other clini-
biopsy in a way that is cost-effective and noninvasive. The cal abnormalities—elevated blood urea nitrogen (BUN) and
urine is collected in midstream or, if that is not feasible, by serum creatinine levels, abnormal urinary sediment, or
bladder catheterization. The urine should be examined evidence of systemic illness (eg, fever, rash, vasculitis).
within 1 hour after collection to avoid destruction of There are several reasons for development of protein-
formed elements. Urinalysis includes a dipstick examina- uria: (1) Functional proteinuria is a benign process stem-
tion followed by microscopic assessment if the dipstick has ming from stressors such as acute illness, exercise, and
positive findings. The dipstick examination measures uri- “orthostatic proteinuria.” The latter condition, generally
nary pH, protein, hemoglobin, glucose, ketones, bilirubin, found in people under age 30 years, usually results in uri-
nitrites, and leukocyte esterase. Urinary specific gravity is nary protein excretion of < 1 g/d. The orthostatic nature of
often reported. Microscopy provides examination of the proteinuria is confirmed by measuring an 8-hour

www.ketabpezeshki.com 66485457-66963820
CMDT15_CH22-p0890-p0929.indd 890 7/10/14 10:54 AM

Potrebbero piacerti anche