ArticlePDF AvailableLiterature Review

Lithium-induced nephrogenic diabetes insipidus: New clinical and experimental findings

Authors:

Abstract and Figures

Lithium (Li+) salts are widely used to treat bipolar mood disorders. Recent trials suggest a potential efficacy also in the treatment of amyotrophic lateral sclerosis and Alzheimer's disease. Li+ is freely filtered by the glomerulus and mainly reabsorbed in the proximal convoluted tubule. Reabsorption in the distal nephron becomes significant under sodium-restricted conditions. Nevertheless, the distal nephron is greatly affected by Li+ even under normal sodium intake. Polyuria, renal tubular acidosis and finally chronic renal failure are the most frequent adverse effects. The occurrence of an overt nephrogenic diabetes insipidus (NDI) limits Li+ usage and imposes suspension. The molecular mechanisms of Li+-related urinary concentration defect involve a dysregulation of the aquaporin system in principal cells of the collecting duct. ENaC is crucial as the entry route for intracellular Li+ accumulation. The basolateral exit route is not clearly identified, but some evidence suggests Na+/H+ exchanger 1 (NHE1) as a potential candidate. Li+ promotes polyuria mainly counteracting the intracellular vasopressin signaling. An additional role of the inner medullary interstitial cells and PGE-2 pathway has to be considered. The GSK3ß cascade is also regulated by Li+. GSK3ß inhibition could lead not only to the polyuria, but also to the Li+-dependent proliferative effect on principal cells. Cellular reorganization of the collecting duct and microcysts are the main pathological findings during Li+ treatment. Their relationship with the urinary concentration defect and an eventual Li+-induced ciliopathy has to been investigated. Li+-induced NDI has been a matter of investigation since the early 1970s. This manuscript reports the latest clinical and experimental findings in combination with the older fundamental results.
Content may be subject to copyright.
S43
www.sin-italy.org/jnonline – www.jnephrol.com
© 2010 Società Italiana di Nefrologia - ISSN 1121-8428
EPHROL
JN 2010; 23 (S16): S43-S48
In t r o d u c t I o n
Lithium (Li+) salts are the first-line therapy for bipolar mood
disorders (1). Li+ has been widely used for mood disorders
since 1970. The development of novel narcoleptics has
decreased the use of Li+ salts. Today a new potential effi-
cacy of lithium in lowering the progression of amyotrophic
lateral sclerosis (2) and Alzheimer’s disease (3) is attract-
ing the attention of clinicians and researchers.
About 50% of patients under lithium treatment experi-
ence urinary concentration defects (4). Therefore, it has
been reported that long-term lithium therapy (more than
10-20 years) can induce chronic kidney failure promot-
ing a tubulointerstitial nephritis with a slow progression to
end-stage renal disease (5). This condition is irreversible.
Other reports showed an incomplete form of renal tubular
acidosis in patients taking lithium (6).
Several experimental models showed that dysregulation of
aquaporins in the collecting duct (CD) is a crucial mecha-
nism for the development of Li+-dependent nephrogenic di-
abetes insipidus (NDI). Li+ affects many regulatory pathways
counteracting the vasopressin-dependent cellular signaling
and also rearranging the entire cellular structure of the CD.
Although Li+ is a very efficacious drug for psychiatrics, re-
nal adverse effects are a limiting problem. This manuscript
reviews the latest clinical and experimental studies on the
Li+-induced urinary concentration impairment.
re n a l h a n d l I n g o f l I t h I u m
Li+ is a monovalent cation that shares some features with
more abundant cations present in the body (Na+ and Mg++).
Li+ can take the place of Mg++, competitively, inhibiting the
activity of GSK3β (7). In addition, in vitro evidence shows
that Li+ can compete with Na+ for the amiloride-sensitive
epithelial Na+ channel (ENaC), Na+/H+ exchanger 3 (NHE3)
and Na+/K+/2Cl- (NKCC2) cotransporter (8, 9).
ab s t r a c t
Lithium (Li+) salts are widely used to treat bipolar mood
disorders. Recent trials suggest a potential efficacy also
in the treatment of amyotrophic lateral sclerosis and Al-
zheimer’s disease. Li+ is freely filtered by the glomerulus
and mainly reabsorbed in the proximal convoluted tu-
bule. Reabsorption in the distal nephron becomes signif-
icant under sodium-restricted conditions. Nevertheless,
the distal nephron is greatly affected by Li+ even under
normal sodium intake. Polyuria, renal tubular acidosis
and finally chronic renal failure are the most frequent ad-
verse effects. The occurrence of an overt nephrogenic
diabetes insipidus (NDI) limits Li+ usage and imposes
suspension. The molecular mechanisms of Li+-related
urinary concentration defect involve a dysregulation of
the aquaporin system in principal cells of the collecting
duct. ENaC is crucial as the entry route for intracellular
Li+ accumulation. The basolateral exit route is not clearly
identified, but some evidence suggests Na+/H+ exchang-
er 1 (NHE1) as a potential candidate.
Li+ promotes polyuria mainly counteracting the intracel-
lular vasopressin signaling. An additional role of the inner
medullary interstitial cells and PGE-2 pathway has to be
considered. The GSK3β cascade is also regulated by Li+.
GSK3β inhibition could lead not only to the polyuria, but
also to the Li+-dependent proliferative effect on principal
cells. Cellular reorganization of the collecting duct and
microcysts are the main pathological findings during Li+
treatment. Their relationship with the urinary concentra-
tion defect and an eventual Li+-induced ciliopathy has to
been investigated. Li+-induced NDI has been a matter
of investigation since the early 1970s. This manuscript
reports the latest clinical and experimental findings in
combination with the older fundamental results.
Key words: Lithium, Nephrogenic diabetes insipidus,
Collecting duct
Department of Anatomy, The Water and Salt Research
Center, Aarhus University, Aarhus C - Denmark
Francesco Trepiccione, Birgitte Mønster Christensen
Lithium-induced nephrogenic diabetes insipidus:
new clinical and experimental findings
ACID-BASE BALANCE: BASIC ASPECTS
S44
Trepiccione and Mønster Christensen: Lithium-induced urinary concentrating defect
Endogenous level of Li+ in healthy humans are quite low,
ranging around 0.2 μmol/L (10). Renal handling of Li+, in
the therapeutic range, is similar to the sodium (Fig. 1). Li+
does not have a high affinity to serum proteins, and it is
freely filtered by the glomerulus. Micropuncture studies
have shown that 65%-75% of the filtered amount is reab-
sorbed along the proximal tubule (PT) mainly in a paracel-
lular transport. One third of this transport occurs in the
straight part of the PT. Proximal Li+ and Na+ reabsorption
are proportional (11). Neither a significant distal absorp-
tion nor tubular secretion mechanisms are described un-
der normal sodium intake in humans. These features made
Li+ clearance an easy tool for estimating sodium and water
reabsorption in the proximal tubule under normal sodium
intake (12). However, clinical (13) and experimental (14)
studies have shown that Li+ is reabsorbed mainly through
a transepithelial route along the thick ascending limb. This
mechanism accounts for about the 20% of the Li+ filtered
amount in salt-depleted subjects (13).
Experimental studies illustrated that a low-sodium diet in-
duces distal Li+ reabsorption (15). Amiloride (16) but not
thiazides (17) can revert this phenomenon, pointing to the
CD as the crucial segment of Li+ distal reabsorption. Recent
Fig. 1 - Renal handling of lithium along the nephron: 65%-
75% of the filtered amount is reabsorbed along the proximal
tubule, mainly in the straight part (large gray arrow). This
reabsorption is paracellular. In salt-depleted status, Li reab-
sorption along the thick ascending limb of Henle’s loop is es-
timated around 20% (small arrow). In rats, a small amount of
lithium is taken up along the amiloride-sensitive distal part of
the nephron (arrowhead).
studies showed that Li+ is taken up from the ENaC to the
same extent as Na+, and this could be relevant for the Li+-
induced urinary concentrating defect.
lI+-I n d u c e d n e p h r o g e n I c d I a b e t e s I n s I p I d u s :
c l I n I c a l d a t a
Li+ therapy can progressively reduce urinary concentration
ability according to the length of the exposure and the dos-
ing. A single dose (600 mg) of Li+-carbonate impairs the an-
tidiuretic response of hypertonic saline infusion in healthy
subjects (18). One month of Li+ treatment reduces both
urinary osmolality and aquaporin 2 excretion in healthy vol-
unteers (19). Longer treatment can progressively induce a
more severe impairment of the urinary concentration ability
up to an overt polyuria (about 10 l/day). Patients with Li+-
induced polyuria have higher levels of circulating vasopres-
sin than healthy controls (20). Finally, Li+-associated polyu-
ria does not respond to deamino-D-vasopressin (dDAVP)
administration (21). This shows a picture of a Li+-induced
NDI. The therapeutic range of lithium is quite narrow. Li+-
related renal adverse effects are also relative to dosages. A
recent review illustrates that a single daily dose is associ-
ated with a lower incidence of renal adverse effects than a
multiple daily dose (22).
Alterations in urinary concentration usually occur after 8
weeks of therapy (23). A drug washout progressively im-
proves the symptoms; they revert progressively up to 8
weeks from after the Li+ withdrawal has started (24). This
reversal seems to be dependent on the time of Li+ exposure,
and is lacking in subjects exposed for more than 15 years
(25). More evidence is necessary to clarify when and how
the NDI symptoms become irreversible with Li+ withdrawal.
Many studies from the 1970s have investigated the histolog-
ical features of patients under Li+ therapy. The distal tubule
and CD are dilated, and their cells show a glycogen-con-
taining PAS-positive material (26). The severity of the tubular
abnormalities positively correlate with urinary concentration
impairment (27). Li+ treatment also induces microcyst gen-
eration. They are easily detected by magnetic resonance
imaging (MRI) scan to be located in the cortex and medulla
and usually ranging around 1-2 mm in diameter (28). Micro-
cysts correlate with the duration of Li+ therapy (26).
Reduction or suspension of treatment are the first steps in
controlling the polyuria; nevertheless, some drugs are ef-
ficacious. Amiloride has been used for a long time in the
treatment of Li+-induced NDI. It was first introduced for
the prevention of the potassium loss associated with an
overt polyuria. Amiloride improves the concentrating ability
of Li+ NDI patients regardless of any significant changes
S45
EPHROL
JN 2010; 23 (S16): S43-S48
in serum Li+ concentration (29). A recent study has shown
that amiloride treatment increases the response to dDAVP
in terms of urinary osmolality, and AQP2 excretion (30). In-
domethacin and other nonsteroidal antiinflammatory drugs
(NSAIDs) have been used for the management of severe
Li+-induced polyuria (31). Short-time usage of NSAIDs is ef-
fective in decreasing the urinary output, but more exhaus-
tive data are necessary on their renal safety in long-term
trials. Whether the pharmacological therapy also has a role
in improving the Li+-induced morphological changes has to
be investigated.
lI+-I n d u c e d n e p h r o g e n I c d I a b e t e s I n s I p I d u s :
e x p e r I m e n t a l d a t a
In vitro and in vivo studies showed that Li+ decreases AQP2
expression (Fig. 2). The vasopressin-regulated water chan-
nel AQP3 is also down-regulated by Li+ treatment (32). Li+
determines a vasopressin nonresponding form of diabetes
insipidus down-regulating luminal AQP2 expression and ba-
solateral AQP3 in principal cells.
The ENaC is the entry route of Li+ in principal cells. Li+ fails to
induce NDI in mice lacking ENaCs in CDs (33). In addition,
amiloride treatment significantly reduces the Li+-induced
polyuria (34). No clear evidence exists for the exit pathways
of Li+. NHE1 in erythrocytes is crucial for Na-Li countertrans-
port (35); this protein could play a similar role in CDs.
Many intracellular pathways can be involved in this process.
Impairment of protein kinases A (PKA) cascade has been
considered the main mechanism of Li+ action. Li+ inhibits
both basal and dDAVP-stimulated activity of adenylate cy-
clase enzyme, thereby reducing the 3’-5’ cyclic adenosine
monophosphate (cAMP) production in isolated CDs from
Li+-treated rats (36). It is controversial whether this mecha-
nism is directly related to AQP2 down-regulation. Mpk-CCD
cells treated with Li+ showed a non-cAMP-dependent down-
regulation of AQP2 (37). A recent study depicts another sce-
nario. Selective GSK3β deletion in CDs is associated with
a lower activity of adenylate cyclase and lower cAMP level,
suggesting a GSK3β-dependent regulation of adenylate cy-
clase (38).
Besides cAMP-dependent pathways, several other key
enzymes are regulated by Li+. Phospho-proteomic analy-
sis on inner medullary CDs from long-term Li+-treated rats
showed Li+ activates PKB/Akt and MAPK kinases and in-
hibits GSK3β kinase (39). Akt is an upstream regulator of
GSK3β. Among several functions, this pathway regulates
the proapoptotic signals. Li+ activates Akt by promoting its
phosphorylation at S473 and T308. Therefore Akt inhibits
GSK3β through phosphorylation at S9. GSK3β inhibition is
associated with an accumulation of β-catenin in principal
cells. This latter protein is mainly involved in cadherin-me-
diated cell adhesion, and it plays a role as transcriptional
coregulator of cell growth genes. This regulatory pathway
Fig. 2 - The collecting duct’s principal cells.
“A”: principal cells are equipped with epi-
thelial Na+ channel (ENaC) on the luminal
side and Na+/H+ exchanger 1 (NHE-1) at
the basolateral side. They are considered
the entry and exit route of Li+ into and out
of the cell, respectively. “B”: Li+ impairs
the PKA pathways. Li+ inhibits adenylate
cyclase activity (AC) and cAMP genera-
tion. “C”: Li+ induces phosphorylation of
the Akt at S473 and T308. This leads to an
inhibition of GSK3β through phosphoryla-
tion at S9. The Akt-GSK pathway can in-
fluence the regulation of AQP2 at several
levels. Recent evidence showed a lower
activity of AC in mice with collecting duct
(CD)-selective knockout for GSK3β. “D”:
role of interstitial cells in AQP2 regulation.
Li+ inhibition of GSK3β activity in intersti-
tial cells stimulates COX2-PGE pathway
leading to AQP2 down-regulation in prin-
cipal cells.
S46
Trepiccione and Mønster Christensen: Lithium-induced urinary concentrating defect
could potentially lead to the high proliferation rate of prin-
cipal cells during long-term Li+ treatment (40). In addition, a
recent study showed a vasopressin-dependent regulation
of the phosphorylation status of β-catenin (41). Whether Li+
counteracts the vasopressin action also by modulating the
phosphorylation status of β-catenin is an interesting future
direction deserving a deeper investigation. Finally Li+-in-
duced NDI pathogenesis involves, at least partly, the med-
ullary interstitial cells. Li+ inhibits the GSK3β and increases
the expression of COX-2 in medullary interstitial cells. This
is associated with an increased urinary excretion of PGE-2
and to the diuretic effect. COX-2 inhibitors partially reverse
Li+-induced polyuria in rats (42). Higher sensitivity to puri-
nergic stimuli could be one of the driving forces of PGE-2
secretion (43).
Li+ remodels the entire structure of the CD. The usual ratio
between principal and intercalated cells (ICs) forming the
CD is subverted after long-term Li+ treatment along all of
the CD zones. The fraction of type A ICs is increased. Type
A ICs appear in the inner medulla, where they are usually
rare (44). Despite this cellular reorganization, principal cells
have a higher proliferation rate than type A ICs (40). Cellular
conversion of principal into type A ICs could be one of the
possible underlying mechanisms (unpublished data).
Both in humans and rats, Li+ treatment is associated with
microcyst formation. Many studies have shown that abnor-
mal renal cyst formation is related to ciliopathies (impair-
ment of primary cilium complex function). A recent study
illustrates that Li+ induces the elongation of the primary cil-
ium in fibroblast-like synoviocytes in a GSK3β not related
mechanism (45).
co n c l u s I o n a n d p e r s p e c t I v e s
Li+ salts are efficacious antipsychotic agents; however, NDI
is a frequent adverse effect that requires cessation of the
therapy. These renal consequences are mainly due to an
impairment of the aquaporin system in the CD. In patients
affected by Li+-dependent NDI, amiloride improves the uri-
nary concentration ability. Experimental data confirm the
crucial importance of the amiloride-sensitive ENaC as the
entry route for Li+ in the CD principal cells. However, de-
spite the fact that many key components have been iden-
tified, the complete picture is still lacking. The detection
of earlier targets involved in Li+-affected AQP2 pathways
will add new insights into aquaporin physiology and NDI
pathogenesis.
Li+ induces severe alterations of the morphology of the CD.
Human biopsies reveal several degrees of lesion from cell
swelling to necrosis and tubular interstitial nephritis. CDs of
Li+-treated rats show a dysregulation of the ratio between
principal and ICs, but how these pathological alterations
occur and what the underlying mechanisms are have still
been only poorly investigated.
Li+ effects on renal physiology have been investigated for
many years. And, although many points have been ad-
dressed, new intriguing questions have arisen and continue
to take the Li+ story forward.
Financial support: None.
Conflict of interest statement: None declared.
Address for correspondence:
Francesco Trepiccione
Department of Anatomy
The Water and Salt Research Center
Aarhus University
Wilhelm Meyers Allé 3
DK-8000 Aarhus C, Denmark
francesco.trepiccione@ana.au.dk
re f e r e n c e s
1. Lister-Williams RH. Relapse prevention in bipolar disorder:
a critical review of current guidelines. J Psychopharmacol.
2006;20:12-16.
2. Fornai F, Longone P, Cafaro L, et al. Lithium delays progres-
sion of amyotrophic lateral sclerosis. Proc Natl Acad Sci U S
A. 2008;105:2052-2057.
3. Martinez A, Perez DI. GSK-3 inhibitors: a ray of hope for
the treatment of Alzheimer’s disease? J Alzheimers Dis.
S47
EPHROL
JN 2010; 23 (S16): S43-S48
2008;15:181-191.
4. Wallin L, Alling C, Aurell M. Impairment of renal function
in patients on long-term lithium treatment. Clin Nephrol.
1982;18:23-28.
5. Presne C, Fakhouri F, Noel LH, et al. Lithium-induced neph-
ropathy: rate of progression and prognostic factors. Kidney
Int. 2003;64:585-592.
6. Batlle D, Gaviria M, Grupp M, Arruda JA, Wynn J, Kurtzman
NA. Distal nephron function in patients receiving chronic lith-
ium therapy. Kidney Int. 1982;21:477-485.
7. Ryves WJ, Harwood AJ. Lithium inhibits glycogen synthase
kinase-3 by competition for magnesium. Biochem Biophys
Res Commun. 2001;280:720-725.
8. Holstein-Rathlou NH. Lithium transport across biological
membranes. Kidney Int Suppl. 1990;28:S4-S9.
9. Kinne R, Koenig B, Hannafin J, Kinne-Saffran E, Scott DM,
Zierold K. The use of membrane vesicles to study the NaCl/
KCl cotransporter involved in active transepithelial chloride
transport. Pflugers Arch. 1985;405(Suppl 1):S101-S105.
10. Fleishman DG, Solyus AA, Gurevich ZP, Bagrov Y. Lithium in
the human body. Hum Physiol. 1985;11:430-435.
11. Corman B, Roinel N, de Rouffignac C. Dependence of water
movement on sodium transport in kidney proximal tubule: a
microperfusion study substituting lithium for sodium. J Membr
Biol. 1981;62:105-111.
12. Thomsen K. Lithium clearance: a new method for determin-
ing proximal and distal tubular reabsorption of sodium and
water. Nephron. 1984;37:217-223.
13. Atherton JC, Doyle A, Gee A, et al. Lithium clearance:
modification by the loop of Henle in man. J Physiol.
1991;437:377-391.
14. Unwin RJ, Walter SJ, Shirley DG. Lithium reabsorption in
perfused loops of Henle: effects of perfusion rate and bu-
metanide. Am J Physiol. 1994;266:F806-F812.
15. Boer WH, Joles JA, Koomans HA, Dorhout Mees EJ. De-
creased lithium clearance due to distal tubular lithium
reabsorption in sodium-depleted dogs. Ren Physiol.
1987;10:65-68.
16. Kirchner KA. Lithium as a marker for proximal tubular deliv-
ery during low salt intake and diuretic infusion. Am J Physiol.
1987;253:F188-F196.
17. Boer WH, Koomans HA, Dorhout Mees EJ. Acute effects of
thiazides, with and without carbonic anhydrase inhibiting ac-
tivity, on lithium and free water clearance in man. Clin Sci
(Lond). 1989;76:539-545.
18. Pedersen RS, Bentzen H, Bech JN, Pedersen EB. Effect
of an acute oral lithium intake on urinary Aquaporin-2 in
healthy humans with and without simultaneous stimulation
with hypertonic saline infusion. Scand J Clin Lab Invest.
2003;63:181-194.
19. Walker RJ, Weggery S, Bedford JJ, McDonald FJ, Ellis G,
Leader JP. Lithium-induced reduction in urinary concentrat-
ing ability and urinary aquaporin 2 (AQP2) excretion in healthy
volunteers. Kidney Int. 2005;67:291-294.
20. Padfield PL, Park SJ, Morton JJ, Braidwood AE. Plasma
levels of antidiuretic hormone in patients receiving pro-
longed lithium therapy. Br J Psychiatry. 1977;130:144-147.
21. Forrest JN Jr, Cohen AD, Torretti J, Himmelhoch JM, Epstein
FH. On the mechanism of lithium-induced diabetes insipidus
in man and the rat. J Clin Invest. 1974;53:1115-1123.
22. Ljubicic D, Letica-Crepulja M, Vitezic D, Bistrovic IL, Ljubi-
cic R. Lithium treatments: single and multiple daily dosing.
Can J Psychiatry. 2008;53:323-331.
23. Boton R, Gaviria M, Batlle DC. Prevalence, pathogenesis,
and treatment of renal dysfunction associated with chronic
lithium therapy. Am J Kidney Dis. 1987;10:329-345.
24. Bucht G, Wahlin A. Renal concentrating capacity in long-
term lithium treatment and after withdrawal of lithium. Acta
Med Scand. 1980;207:309-314.
25. Bendz H, Sjodin I, Aurell M. Renal function on and off lith-
ium in patients treated with lithium for 15 years or more:
a controlled, prospective lithium-withdrawal study. Nephrol
Dial Transplant. 1996;11:457-460.
26. Walker RG, Escott M, Birchall I, Dowling JP, Kincaid-Smith
P. Chronic progressive renal lesions induced by lithium.
Kidney Int. 1986;29:875-881.
27. Hansen HE, Hestbech J, Sorensen JL, Norgaard K,
Heilskov J, Amdisen A. Chronic interstitial nephropa-
thy in patients on long-term lithium treatment. Q J Med.
1979;48:577-591.
28. Farres MT, Ronco P, Saadoun D, et al. Chronic lithium
nephropathy: MR imaging for diagnosis. Radiology.
2003;229:570-574.
29. Batlle DC, von Riotte AB, Gaviria M, Grupp M. Ameliora-
tion of polyuria by amiloride in patients receiving long-term
lithium therapy. N Engl J Med. 1985;312:408-414.
30. Bedford JJ, Weggery S, Ellis G, et al. Lithium-induced
nephrogenic diabetes insipidus: renal effects of amiloride.
Clin J Am Soc Nephrol. 2008;3:1324-1331.
31. Allen HM, Jackson RL, Winchester MD, Deck LV, Allon M. In-
domethacin in the treatment of lithium-induced nephrogenic
diabetes insipidus. Arch Intern Med. 1989;149:1123-1126.
32. Kwon TH, Laursen UH, Marples D, et al. Altered ex-
pression of renal AQPs and Na(+) transporters in rats
with lithium-induced NDI. Am J Physiol Renal Physiol.
2000;279:F552-F564.
33. Christensen BM, Loffing J, Hummler E, Rossier BC. Collect-
ing duct-specific gene inactivation of alpha ENaC protects
mice against lithium-induced nephrogenic diabetes insipidus
[abstract]. J Am Soc Nephrol. 2006;17:295A.
34. Kortenoeven ML, Li Y, Shaw S, et al. Amiloride blocks lith-
ium entry through the sodium channel thereby attenuating
the resultant nephrogenic diabetes insipidus. Kidney Int.
2009;76:44-53.
S48
Trepiccione and Mønster Christensen: Lithium-induced urinary concentrating defect
35. Zerbini G, Maestroni A, Mangili R, Pozza G. Amiloride-
insensitive Na+-H+ exchange: a candidate mediator of
erythrocyte Na+-Li+ countertransport. J Am Soc Nephrol.
1998;9:2203-2211.
36. Jackson BA, Edwards RM, Dousa TP. Lithium-induced polyu-
ria: effect of lithium on adenylate cyclase and adenosine
3’,5’-monophosphate phosphodiesterase in medullary ascend-
ing limb of Henle’s loop and in medullary collecting tubules. En-
docrinology. 1980;107:1693-1698.
37. Li Y, Shaw S, Kamsteeg EJ, Vandewalle A, Deen PM. Devel-
opment of lithium-induced nephrogenic diabetes insipidus is
dissociated from adenylyl cyclase activity. J Am Soc Nephrol.
2006;17:1063-1072.
38. Kuure S, Popsueva A, Jakobson M, Sainio K, Sariola H. Gly-
cogen synthase kinase-3 inactivation and stabilization of beta-
catenin induce nephron differentiation in isolated mouse and rat
kidney mesenchymes. J Am Soc Nephrol. 2007;18:1130-1139.
39. Nielsen J, Hoffert JD, Knepper MA, Agre P, Nielsen S, Fenton
RA. Proteomic analysis of lithium-induced nephrogenic diabe-
tes insipidus: mechanisms for aquaporin 2 down-regulation and
cellular proliferation. Proc Natl Acad Sci U S A. 2008;105:3634-
3639.
40. Christensen BM, Kim YH, Kwon TH, Nielsen S. Lithium
treatment induces a marked proliferation of primarily princi-
pal cells in rat kidney inner medullary collecting duct. Am J
Physiol Renal Physiol. 2006;291:F39-F48.
41. Bansal AD, Hoffert JD, Pisitkun T, et al. Phosphoproteomic
profiling reveals vasopressin-regulated phosphorylation sites
in collecting duct. J Am Soc Nephrol. 2010;21:303-315.
42. Rao R, Zhang MZ, Zhao M, et al. Lithium treatment in-
hibits renal GSK-3 activity and promotes cyclooxyge-
nase 2-dependent polyuria. Am J Physiol Renal Physiol.
2005;288:F642-F649.
43. Zhang Y, Kohan DE, Nelson RD, Carlson NG, Kishore BK.
Potential involvement of P2Y2 receptor in diuresis of pos-
tobstructive uropathy in rats. Am J Physiol Renal Physiol.
2010;298:F634-F642.
44. Christensen BM, Marples D, Kim YH, Wang W, Frokiaer J,
Nielsen S. Changes in cellular composition of kidney collect-
ing duct cells in rats with lithium-induced NDI. Am J Physiol
Cell Physiol. 2004;286:C952-C964.
45. Ou Y, Ruan Y, Cheng M, Moser JJ, Rattner JB, van der Hoorn
FA. Adenylate cyclase regulates elongation of mammalian
primary cilia. Exp Cell Res. 2009;315:2802-2817.
... In the present patient, NDI did not improve despite discontinuation of lithium and undetectable serum lithium levels. Typically, the loss of urinary concentrating ability induced by lithium manifests after approximately eight weeks of administration and gradually recovers to baseline levels approximately eight weeks after discontinuation (13). The extent of recovery appears to be influenced by the duration of lithium exposure, with reports indicating that the urine concentrating ability fails to recover in individuals using lithium for more than 15 years (9). ...
... The extent of recovery appears to be influenced by the duration of lithium exposure, with reports indicating that the urine concentrating ability fails to recover in individuals using lithium for more than 15 years (9). The dose and duration of lithium use required to cause irreversible NDI remain unclear (13). However, in this case, the patient had been taking lithium for 34 years, suggesting that long-term lithium exposure contributed to the irreversibility of the NDI. ...
... NSAIDs offer a swift reduction in urine output, making them valuable in emergencies. However, the safety of longterm NSAIDs use remains unestablished, and its use should be limited to short-term treatments (13,15). In this case, the patient experienced progressive renal dysfunction, likely attributable to NSAIDs, necessitating discontinuation. ...
Article
Nephrogenic diabetes insipidus (NDI) is characterized by excessive urination and an inability to concentrate urine. Lithium is the most common cause of acquired NDI. Treatment typically involves thiazide diuretics and nonsteroidal anti-inflammatory drugs (NSAIDs). However, the efficacy of desmopressin in NDI remains unclear. We herein report a case of lithium-induced NDI in a 71-year-old woman with lithium-induced NDI. Thiazide diuretics and NSAIDs reduced the urine output by approximately 40% compared to pretreatment, while the addition of desmopressin reduced it by approximately 70%. This case suggests that desmopressin can be a viable treatment option for lithium-induced NDI.
... 5 One of the most frequent renal side effects is NDI, resulting in polydipsia and polyuria. 6,7 NDI has been described in humans and in experimental animal models and develops shortly after the initiation of the treatment. In healthy volunteers, NDI appears 8 days after lithium treatment initiation and resolves shortly after discontinuation. ...
... Median age was 51 (37-62) years, 38% of the individuals were males, median mGFR was 78 (63-90) ml/min/1.73 m 2 , and median lithium treatment duration was 5 (2)(3)(4)(5)(6)(7)(8)(9)(10)(11)(12)(13)(14) years. Median 24-hour urine output was 1932 (1448-2835) ml/ d, and 21% of the patients displayed polyuria defined as urine output >3 l/d. ...
Article
Full-text available
Background and objectives Lithium treatment can induce nephrogenic diabetes insipidus, but no consensus intervention is offered to date. We evaluated in these patients patterns of urine concentration and the correlates of 24-hour urine output. Methods Prospective single-center observational study of 217 consecutive lithium-treated individuals, with 24-hour urine collection, desmopressin (DDAVP) concentrating test, fasting plasma vasopressin measurement (copeptin measurement in n=119), and measured GFR. Maximal urine osmolality (MaxUosm) was the highest level during the DDAVP test. Results 21% of individuals displayed polyuria (> 3l/day), but 55% displayed elevated fasting vasopressin level (>5pg/ml). Uosm was significantly lower, and urinary output and free water clearance were significantly higher in individuals treated more than 10 years. MaxUosm was >600mOsm/KgH2O in 128 patients (59%), among which vasopressin was increased in 51%, associated with higher lithium dose (950 [750-1200] versus 800 [500-1000] mg/d, p<0.001). 100% of patients with lithium daily dose ≥1400 mg/d had high vasopressin levels. In multivariable analysis, 24-hour urine output was associated with higher lithium daily dose (β 0.49 ± 0.17, p=0.005), female sex (β -359 ± 123, p=0.004), daily osmolar intake (β 2.21 ± 0.24, p<0.001), MaxUosm (β -2.89 ± 0.35, p<0.001) and plasma vasopressin level (β 10.17 ± 4.76, p=0.03). Conclusions Higher lithium daily dose was associated with higher vasopressin levels and higher urine output, independently of other factors. Daily osmolar intake was also associated with higher 24-hour urine output. These results suggest that controlled salt and protein intake and lithium dose might reduce polyuria in these patients.
... Приобретенные формы могут быть следствием лечения препаратами, блокирующими действие АДГ, включая литий, некоторые антибиотики, противогрибковые и противоопухолевые средства. К другим факторам относятся заболевания почек, такие как хронический пиелонефрит, интерстициальный нефрит, поликистоз, обструктивная уропатия, почечный амилоидоз, саркоидоз, синдром Барттера и метаболические нарушения -гипокалиемия и гиперкальциемия [3][4][5]. ...
Article
Nephrogenic diabetes insipidus is a heterogeneous disease in the etiopathogenesis of which are involved acquired and congenital factors. In this case, the kidneys do not respond to vasopressin and continue to produce large concentrated volumes of urine. Distinctive features in the pathology of central genesis are the fact of trauma, brain tumor with involvement of pituitary gland, response to intranasal vasopressin in the form of decreased diuresis. For diagnosis it is important to perform a series of differential diagnostic tests. In this article we present a unique clinical case of a patient with a long-term course of diabetes insipidus with the development a spectrum of comorbid conditions, metabolic disorders, required kidney transplantation.
... Chronic lithium treatment has been associated with several different forms of renal side effects. 7,[10][11][12] Nephrogenic diabetes insipidus is the most common renal side effect of lithium therapy 13,14 and has been well investigated. Less is known about potential glomerular toxicity of lithium, particularly the nephrotic syndrome. ...
Article
Background: The use of lithium during breast-feeding has not been comprehensively investigated in humans due to concerns about lithium toxicity. Procedure: We analyzed lithium in the kidneys of nursed pups of lithium medicated mothers, using analytical spectroscopy in a novel rat model. The mothers were healthy rats administered lithium via gavage (1000 mg/day Li2 CO3 per 50 kg body weight). Results: Lithium was detected in the breast milk, and in the blood of pups (0.08 mM), of lithium-exposed dams at post-natal day 18 (P18), during breast-feeding. No lithium was detected after breast-feeding, at P25 (four days after cessation of nursing). The lithium pups blood had elevated urea nitrogen at P18 and reduced total T4 at P18 and P25, indicating a longer-term effect on the kidneys and the thyroid gland. Multivariate machine learning analysis of spectroscopy data collected from the excised kidneys of pups showed elevated potassium in lithium-exposed animals both during- and after breast-feeding. The elevated renal potassium was associated with low Nephrin expression in the kidneys measured immunohistochemically during breast-feeding. After lithium exposure is stopped, the filtration of lithium from the kidneys reverses these effects. Our study showed that breastfeeding during lithium use has an effect on the kidneys of the offspring in rats.
... Lithium is the cornerstone treatment of bipolar disorder, characterized by potentially life-threatening manic and depressive episodes. One of the main adverse effects of this treatment is NDI leading to polyuria and polydipsia, occurring in 20%-70% of patients depending on the population and the definition of NDI [88]. As a monovalent cation, lithium follows sodium transport along the kidney tubule and enters the PC of the CD via ENaC [89]. ...
Article
Full-text available
The kidneys, by equilibrating the outputs to the inputs, are essential for maintaining constant the volume, pH and electrolyte composition of the internal milieu. Inability to do so, either because of internal kidney dysfunction (primary alteration) or because some external factors (secondary alteration) would lead to pathologies, more or less severe, leading to modification of these parameters and affecting the functions of other organs. Alterations of the functions of the collecting duct (CD), the most distal part of the nephron, have been extensively studied and have led to a better diagnosis, better management of the related diseases and the development of therapeutic tools. Thus, dysfunctions of principal cells specific transporters such as ENaC or AQP2 or its receptors (mineralocorticoid or vasopressin receptors) caused by mutations or by compounds present in the environment (lithium, antibiotics…) have been demonstrated in a variety of syndromes (Liddle, pseudohypoaldosteronism type‐1, diabetes insipidus…) affecting salt, potassium and water balance. In parallel, studies of specific transporters (H+‐ATPase, anion exchanger 1) in intercalated cells revealed the mechanisms of related tubulopathies like distal renal distal tubular acidosis or Sjögren syndrome. In this review, we will recapitulate the mechanisms of most of the primary and secondary alteration of the ion transport system of the CD to provide a better understanding of these diseases and highlight how a targeted perturbation may affect many different pathways due to the strong crosstalk and entanglements between the different actors (transporters, cell types). This article is protected by copyright. All rights reserved
... 143 A major proportion of the adults that possess the NDI might have attained the disease with lithium treatment being primarily responsible for it. 144,145,146 The incidence of patients that attain the harmful dangerous implication of lithium treatment could reach up-to 85 %. 146,147 In case of rats, long term lithium doses lasting up to more than one month leads to epithelial remodeling in the collecting duct and a significant reduction in the number of principal cells. 148 In the mpkCCD (mouse collecting duct cell line), lithium exposure reduced Aqp2 expression which is irrespective of the cAMP levels. ...
Article
Full-text available
The expression of various isoforms of aquaporins (AQPs) in different tissues and organs of the body makes it a viable candidate for being responsible for maintaining cell stability and integrity as their involvement has been well documented in a number of pathophysiological conditions of the human body. Any alteration in the cellular environment brought about by these AQPs creates severe downstream effects like changes in cellular osmolality, volume, ionic composition, signaling pathways and even in the levels of intracellular second messengers and, as such, facilitates the occurrence of diseases like cancer. The altered equilibrium of water, extracellular ions and amino acid neurotransmitters caused by neuronal destruction and oxidative stress in neurodegenerative diseases proposed the role of these AQPs in these diseased conditions as well. The association of AQPs in a variety of inflammatory processes like lung injury, brain edema, neuromyelitis optica, and colitis as manifested through their dysregulation both in animal and human diseases is truly an eye opener for their role in protection and reaction to various noxious stimuli including bacterial infection. Renal diseases like nephrogenic diabetes inspidus, autosomal dominant polycystic kidney disease and acute kidney injury are some of the pathophysiological conditions related to malfunctioning of aquaporins. Besides, the malfunctioning of aquaglyceroporins like AQP7 and AQP9 makes them responsible for disorders like obesity, nonalcoholic fatty liver disease and non-alcoholic steatohepatitis. In this review article, we present our current understanding of the role of AQPs in the causation of these metabolic disorders and how targeting them holds promising therapeutic potential for most of these diseases like cancer, renal diseases and even cardiovascular disorders.
Chapter
Despite recent downward trends in lithium use for bipolar disorder (BD) [1], nearly every treatment guideline, meta-analysis or review published in the last decade has reinforced the notion that lithium remains the mood stabilizer of choice for acute or maintenance therapy in those with a history of mania (BD-1, schizoaffective disorder, bipolar type [SAD-BT]), and an important treatment option for other mood disorder spectrum patients (BD-2, unipolar major depressive disorder [MDD]) [2–4].
Chapter
Diabetes insipidus refers to the inability of the kidneys to concentrate the urine by water extraction in the collecting duct. This can be due to a deficiency in the mediating hormone (central diabetes insipidus), variably called antidiuretic hormone (ADH) or arginine vasopressin (AVP), or an impaired ability of the kidneys to respond to the hormone (nephrogenic diabetes insipidus).Affected patients typically present with large urine volumes, typically exceeding 10 L per day in adult patients and necessitating constant water intake. Consequently, patients are at risk of severe hypernatremic dehydration, especially when oral intake is impaired. Whereas central diabetes insipidus can be effectively treated with hormone replacement, management of the nephrogenic form is more difficult and the polyuria can only be ameliorated.Both forms can be either inherited or acquired. Identification of the genes involved in diabetes insipidus has provided tremendous insight into the physiology of urinary concentration.KeywordsDiabetes insipidusAquaporineArginine vasopressinAVPAntidiuretic hormoneADHVasopressin receptorAVPR2Prostaglandin
Article
Full-text available
Central diabetes insipidus (CDI) is a rare endocrine disease deriving from an insufficient production or secretion of anti-diuretic hormone. Recently, CDI has been reported as a rare side effect triggered by immune checkpoint inhibitors (ICI) in cancer patients. Despite its current rarity, CDI triggered by ICI is expected to affect an increasing number of patients because of the expanding use of these effective drugs in a growing number of solid and hematologic malignancies. An appropriate assessment of the severity of adverse events induced by anticancer agents is crucial in their management, including dosing adjustment and temporary withdrawal or discontinuation treatment. However, assessment of the severity of CDI induced by ICI may be challenging, as its main signs and symptoms (polyuria, dehydration, weight loss, and hypernatremia) can be incompletely graded. Indeed, the current grading system of toxicity induced by anticancer treatments does not include polyuria. Additionally, dehydration in patients affected by diabetes insipidus, including ICI-induced CDI, is different in certain aspects from that due to other conditions seen in cancer patients, such as vomiting and diarrhea. This prompted us to reflect on the need to grade polyuria, and how to grade it, and to consider a specific grading system for dehydration associated with CDI induced by ICI. Here we propose a new grading system for polyuria and dehydration, as critical symptoms of the CDI syndrome occurring in patients on ICI treatment, to obtain better management of both the adverse event and the triggering drugs.
Article
Full-text available
Lithium therapy frequently induces nephrogenic diabetes insipidus; amiloride appears to prevent its occurrence in some clinical cases. Amiloride blocks the epithelial sodium channel (ENaC) located in the apical membrane of principal cells; hence one possibility is that ENaC is the main entry site for lithium and the beneficial effect of amiloride may be through inhibiting lithium entry. Using a mouse collecting duct cell line, we found that vasopressin caused an increase in Aquaporin 2 (AQP2) expression which was reduced by clinically relevant lithium concentrations similar to what is seen with in vivo models of this disease. Further amiloride or benzamil administration prevented this lithium-induced downregulation of AQP2. Amiloride reduced transcellular lithium transport, intracellular lithium concentration, and lithium-induced inactivation of glycogen synthase kinase 3beta. Treatment of rats with lithium downregulated AQP2 expression, reduced the principal-to-intercalated cell ratio, and caused polyuria, while simultaneous administration of amiloride attenuated all these changes. These results show that ENaC is the major entry site for lithium in principal cells both in vitro and in vivo. Blocking lithium entry with amiloride attenuates lithium-induced diabetes insipidus, thus providing a rationale for its use in treating this disorder.
Article
Full-text available
Alzheimer's disease (AD) is the most common form of dementia affecting more than 15 millions individuals worldwide. While the cause is unknown, there are two major neuropathological abnormalities present in the brains of patients with AD, the extracellular senile plaques and the intracellular neurofibrillary tangles. There is strong evidence that glycogen synthase kinase-3 (GSK-3) plays an important role in AD being involved in the regulation of these neuropathological hallmarks. Increased activity and/or overexpression of this enzyme in AD is associated with increased tau hyperphosphorylation and alterations in amyloid-beta processing that are thought to precede the formation of neurofibrillary tangles and senile plaques, respectively. Furthermore, over activity of GSK-3 is also involved in neuronal loss. These data clearly identify GSK-3 inhibitors as one of the most promising new approaches for the future treatment of AD and a reduction of the aberrant over activity of this enzyme might decrease several aspects of the neuronal pathology in AD. In this review, we provide an overview of the rationale for the development of GSK-3 inhibitors for the treatment of AD, discussing the risks and benefits of this approach.
Article
Protein phosphorylation is an important component of vasopressin signaling in the renal collecting duct, but the database of known phosphoproteins is incomplete. We used tandem mass spectrometry to identify vasopressin-regulated phosphorylation events in isolated rat inner medullary collecting duct (IMCD) suspensions. Using multiple search algorithms to identify the phosphopeptides from spectral data, we expanded the size of the existing collecting duct phosphoproteome database from 367 to 1187 entries. Label-free quantification in vasopressin- and vehicle-treated samples detected a significant change in the phosphorylation of 29 of 530 quantified phosphopeptides. The targets include important structural, regulatory, and transporter proteins. The vasopressin-regulated sites included two known sites (Ser-486 and Ser-499) present in the urea channel UT-A1 and one previously unknown site (Ser-84) on vasopressin-sensitive urea channels UT-A1 and UT-A3. In vitro assays using synthetic peptides showed that purified protein kinase A (PKA) could phosphorylate all three sites, and immunoblotting confirmed the PKA dependence of Ser-84 and Ser-486 phosphorylation. These results expand the known list of collecting duct phosphoproteins and highlight the utility of targeted phosphoproteomic approaches.
Article
AVP resistance of the medullary collecting duct (mCD) in postobstructive uropathy (POU) has been attributed to increased production of PGE2. P2Y2 receptor activation causes production of PGE2 by the mCD. We hypothesize that increased P2Y2 receptor expression and/or activity may contribute to the diuresis of POU. Sprague-Dawley rats were subjected to bilateral ureteral obstruction for 24 h followed by release (BUO/R, n = 17) or sham operation (SHM/O, n = 15) and euthanized after 1 wk or 12 days. BUO/R rats developed significant polydipsia, polyuria, urinary concentration defect, and increased urinary PGE2 and decreased aquaporin-2 protein abundance in the inner medulla compared with SHM/O rats. After BUO/R, the relative mRNA expression of P2Y2 and P2Y6 receptors was increased by 2.7- and 4.9-fold, respectively, without significant changes in mRNA expression of P2Y1 or P2Y4 receptor. This was associated with a significant 3.5-fold higher protein abundance of the P2Y2 receptor in BUO/R than SHM/O rats. When freshly isolated mCD fractions were challenged with different types of nucleotides (ATPgammaS, ADP, UTP, or UDP), BUO/R and SHM/O rats responded to only ATPgammaS and UTP and released PGE2, consistent with involvement of the P2Y2, but not P2Y6, receptor. ATPgammaS- or UTP-stimulated increases in PGE2 were much higher in BUO/R (3.20- and 2.28-fold, respectively, vs. vehicle controls) than SHM/O (1.68- and 1.30-fold, respectively, vs. vehicle controls) rats. In addition, there were significant 2.4- and 2.1-fold increases in relative mRNA expression of prostanoid EP1 and EP3 receptors, respectively, in the inner medulla of BUO/R vs. SHM/O rats. Taken together, these data suggest that increased production of PGE2 by the mCD in POU may be due to increased expression and activity of the P2Y2 receptor. Increased mRNA expression of EP1 and EP3 receptors in POU may also help accentuate PGE2-induced signaling in the mCD.
Article
The primary cilium is a non-motile microtubule-based structure that shares many similarities with the structures of flagella and motile cilia. It is well known that the length of flagella is under stringent control, but it is not known whether this is true for primary cilia. In this study, we found that the length of primary cilia in fibroblast-like synoviocytes, either in log phase culture or in quiescent state, was confined within a range. However, when lithium was added to the culture to a final concentration of 100 mM, primary cilia of synoviocytes grew beyond this range, elongating to a length that was on average approximately 3 times the length of untreated cilia. Lithium is a drug approved for treating bipolar disorder. We dissected the molecular targets of this drug, and observed that inhibition of adenylate cyclase III (ACIII) by specific inhibitors mimicked the effects of lithium on primary cilium elongation. Inhibition of GSK-3beta by four different inhibitors did not induce primary cilia elongation. ACIII was found in primary cilia of a variety of cell types, and lithium treatment of these cell types led to their cilium elongation. Further, we demonstrate that different cell types displayed distinct sensitivities to the lithium treatment. However, in all cases examined primary cilia elongated as a result of lithium treatment. In particular, two neuronal cell types, rat PC-12 adrenal medulla cells and human astrocytes, developed long primary cilia when lithium was used at or close to the therapeutic relevant concentration (1-2 mM). These results suggest that the length of primary cilia is controlled, at least in part, by the ACIII-cAMP signaling pathway.
Article
One-hundred and ten patients treated with lithium for more than six months were studied in order to determine the prevalence of lithium induced nephropathy. Eighteen of 69 patients (26 per cent) who had been treated for more than two years presented a chronic interstitial nephropathy characterized by a marked decrease in renal concentrating ability with a disproportionate preservation of glomerular filtration rate. Histologically, increased amounts of fibrotic tissue in the medulla and the cortex were found together with tubular atrophy. In 40 per cent of the patients who underwent renal biopsy, cystic formations in the cortex were found. The impairment of renal concentrating ability could be related to the duration of lithium treatment and the degree of tubular damage correlated with the degree of impairment of renal concentrating ability. Lithium induced, chronic nephropathy is a rather common complication of long-term lithium treatment and reduces the patients capacity to regulate water and electrolyte metabolism. As water and electrolyte loss appears to precede the slowly progressing lithium intoxication, the main hazard of lithium induced nephropathy is lithium intoxication.
Article
Among 18 patients receiving prophylactic, long-term lithium treatment for manic-depressive psychosis, a high incidence of increased thirst and frequency of micturition (60–70 per cent) was noted on direct questioning. Symptoms arose at varying times after the start of lithium therapy; in no patient did symptoms antedate the use of the drug. Plasma levels of antidiuretic hormone were found, on average, to be higher than in normal control subjects for a given level of plasma osmolality, although the scatter of results was wide. It is suggested that elevation of antidiuretic hormone occurs as a compensatory mechanism for the polyuria which is a common feature of long-term lithium treatment. The more florid form of nephrogenic diabetes insipidus occasionally seen in lithium takers seems likely to be due to a different mechanism from the more common mild polyuria.
Article
1. The contribution of Li+ reabsorption in the loop of Henle to lithium clearance (CLi) and the possible mechanism(s) involved were assessed in healthy volunteers. Four mechanisms were considered: (a) passive reabsorption in the thin ascending limb, (b) solvent drag in the thin descending limb, (c) the Na+, K+, 2Cl- transporter in the thick ascending limb and (d) paracellular movement in the thick ascending limb. 2. Since alterations in the corticomedullary osmolal concentration gradient produced by fluid restriction (500 ml day-1) and subsequent water loading (15 ml kg-1) did not affect either CLi (28.5 +/- 2.1 vs. 28.2 +/- 1.9 ml min-1) or fractional lithium clearance (FELi; 23.5 +/- 2.0 vs. 23.0 +/- 1.9%), it is unlikely that substantial Li+ reabsorption occurs in the thin limbs by either passive movement or solvent drag. 3. Increasing plasma Li+ with unchanged plasma Na+ in salt-replete volunteers was associated with only small reductions in CLi (32.8 +/- 1.3 ml min-1, P less than 0.05) and FELi (27.3 +/- 1.8 vs. 25.3 +/- 2.0%, P less than 0.05). This suggests that substantial Li+ reabsorption on the Na+, K+, 2Cl- transporter does not occur. 4. Bumetanide increased FELi in salt-depleted (LS) and salt-replete (HS) volunteers and abolished the pre-diuretic difference in FELi between salt intakes (LS, 16.6 +/- 1.5 vs. 38.7 +/- 2.3%, P less than 0.001; HS, 30.1 +/- 1.5 vs. 40.5 +/- 2.0%, P less than 0.001). Changes in CPO4 and CHCO3 were not detected. Acetazolamide produced comparable increases in FELi (LS, 16.6 +/- 1.5 vs. 38.7 +/- 2.2%, P less than 0.001; HS, 30.1 +/- 1.5 vs. 43.1 +/- 2.4%, P less than 0.01); and CPO4 and CHCO3 were increased. When tubular flow to the loop of Henle was increased by acetazolamide, the bumetanide-induced increases in FELi were reduced (LS, 38.7 +/- 2.2 vs. 48.7 +/- 2.3%, P less than 0.001; HS, 43.1 +/- 2.4 vs. 48.1 +/- 2.6%, P less than 0.001). 5. These data are consistent with the view that (a) Li+ is reabsorbed by a bumetanide-sensitive mechanism in the loop of Henle, (b) approximately 20 and 10% of the filtered load, respectively, is reabsorbed in the loop in salt-depleted and salt-replete volunteers, (c) flow-dependent, voltage-driven paracellular movement in the thick ascending limb is the likely mechanism and (d) this mechanism could account for the difference in Li+ reabsorption between low and high salt intakes.
Article
Li+ is actively transported out of cells, and across different epithelia of both mammalian and amphibian origin. Due to the low affinity of the Na+/K(+)-ATPase for Li+, the transport is most likely energized by exchange and/or cotransport processes. The detailed mechanism by which Li+ is reabsorbed across the proximal tubule is not known, although it seems reasonable to assume that at least a part is by secondary active transcellular transport. The evidence further suggest that aldosterone and maybe vasopressin, through their effects on the Na+ channels in the late distal tubule and the collecting duct may be of significance in inducing distal Li+ reabsorption, as seen during severe sodium restriction in rats and dogs. Clearly more studies are needed to finally resolve these issues.