bariatric surgery and effects on calcium and bone metabolism

12
ORIGINAL PAPER Bariatric Surgery and Effects on Calcium and Bone Metabolism Khashayar Sakhaee Ó Springer Science+Business Media New York 2013 Abstract With the increasing epidemic of obesity in the United States as well as abroad, bariatric surgery has emerged as the most effective and sustained treatment for reduction. This treatment modality has been well recog- nized to diminish the risk of cardiovascular morbidity and mortality and ameliorate diabetes mellitus. However, with time, derangement in mineral metabolism has emerged as a major complication in this population. Population-based study has shown increased prevalence of bone fractures and kidney stone formation following bariatric surgery. The risk appears to be more specific after Roux-en-Y gastric bypass procedures, the most common surgical approach among this population. Over the past decade, there have been advances in the understanding of pathophysiologic mechanisms of both bone loss and kidney stone disease in these patients. The understanding of these underlying pathophysiologic mechanisms may lead to the development of drug therapies that ameliorate this complication. Unfortunately, at the present time, there is no hard data on any specific treatment showing decreased incidence of fragility fractures or kidney stone passage. However, some studies suggest that calcium and vitamin D supplementation may decrease bone loss and bone turnover, and as a result, increase bone mineral density in this population. However, there is concern with the development of kidney stone formation following such an approach. A novel treatment approach would be the use of effervescent potassium calcium citrate that not only pre- vents complications of bone loss but may diminish the risk of kidney stone formation. Despite preliminary results showing the effectiveness of this drug in the reduction in the parathyroid hormone, bone turnover, and improvement in the urinary saturation marker showing effectiveness against calcium oxalate and uric acid stones, there is no hard data available to support the effectiveness of this treatment in the reduction in fragility fractures or kidney stone incidence. Such studies to explore this effect must be considered in the future. Keywords Bariatric surgery Á Roux-en-Y Á Kidney stones Á Bone loss Introduction Bariatric surgery has been shown to induce sustained weight loss, thereby significantly reducing morbidity and mortality related to obesity [13]. With the epidemic of overweight and obese populations [4], there has been growing interest in medical and surgical management of obesity. Lifestyle modification and medical therapy induce short-term weight loss with difficult long-term mainte- nance [5, 6]. As a result, an increasing number of surgical operations have been performed in the United States [2, 5, 7]. Previous surgical operations, such as Jejunoileal bypass and biliopancreatic diversions (BPD), have been either abandoned or diminished due to severe morbidity and mortality [811]. In recent years, Roux-en-Y gastric bypass (RYGB) and gastric banding have been promoted [12]. Although these procedures have been successful and salutary in reducing the complications of obesity [6, 13, 14], skeletal bone disease [1520] and nephrolithiasis [2126] have emerged as the main complications of RYGB. K. Sakhaee (&) Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, TX 75390-8885, USA e-mail: [email protected] 123 Clinic Rev Bone Miner Metab DOI 10.1007/s12018-013-9145-2

Upload: khashayar

Post on 20-Dec-2016

215 views

Category:

Documents


3 download

TRANSCRIPT

ORIGINAL PAPER

Bariatric Surgery and Effects on Calcium and Bone Metabolism

Khashayar Sakhaee

� Springer Science+Business Media New York 2013

Abstract With the increasing epidemic of obesity in the

United States as well as abroad, bariatric surgery has

emerged as the most effective and sustained treatment for

reduction. This treatment modality has been well recog-

nized to diminish the risk of cardiovascular morbidity and

mortality and ameliorate diabetes mellitus. However, with

time, derangement in mineral metabolism has emerged as a

major complication in this population. Population-based

study has shown increased prevalence of bone fractures and

kidney stone formation following bariatric surgery. The risk

appears to be more specific after Roux-en-Y gastric bypass

procedures, the most common surgical approach among this

population. Over the past decade, there have been advances

in the understanding of pathophysiologic mechanisms of

both bone loss and kidney stone disease in these patients.

The understanding of these underlying pathophysiologic

mechanisms may lead to the development of drug therapies

that ameliorate this complication. Unfortunately, at the

present time, there is no hard data on any specific treatment

showing decreased incidence of fragility fractures or kidney

stone passage. However, some studies suggest that calcium

and vitamin D supplementation may decrease bone loss and

bone turnover, and as a result, increase bone mineral density

in this population. However, there is concern with the

development of kidney stone formation following such an

approach. A novel treatment approach would be the use of

effervescent potassium calcium citrate that not only pre-

vents complications of bone loss but may diminish the risk

of kidney stone formation. Despite preliminary results

showing the effectiveness of this drug in the reduction in the

parathyroid hormone, bone turnover, and improvement in

the urinary saturation marker showing effectiveness against

calcium oxalate and uric acid stones, there is no hard data

available to support the effectiveness of this treatment in the

reduction in fragility fractures or kidney stone incidence.

Such studies to explore this effect must be considered in the

future.

Keywords Bariatric surgery � Roux-en-Y � Kidney

stones � Bone loss

Introduction

Bariatric surgery has been shown to induce sustained

weight loss, thereby significantly reducing morbidity and

mortality related to obesity [1–3]. With the epidemic of

overweight and obese populations [4], there has been

growing interest in medical and surgical management of

obesity. Lifestyle modification and medical therapy induce

short-term weight loss with difficult long-term mainte-

nance [5, 6]. As a result, an increasing number of surgical

operations have been performed in the United States [2, 5,

7]. Previous surgical operations, such as Jejunoileal bypass

and biliopancreatic diversions (BPD), have been either

abandoned or diminished due to severe morbidity and

mortality [8–11].

In recent years, Roux-en-Y gastric bypass (RYGB) and

gastric banding have been promoted [12]. Although these

procedures have been successful and salutary in reducing

the complications of obesity [6, 13, 14], skeletal bone

disease [15–20] and nephrolithiasis [21–26] have emerged

as the main complications of RYGB.

K. Sakhaee (&)

Department of Internal Medicine, University of Texas

Southwestern Medical Center at Dallas, 5323 Harry Hines

Boulevard, Dallas, TX 75390-8885, USA

e-mail: [email protected]

123

Clinic Rev Bone Miner Metab

DOI 10.1007/s12018-013-9145-2

Prevalence of Bone Fracture and Changes in Bone

Mineral Density Following Bariatric Surgery

Bone Changes Post-RYGB

Despite the abundance of studies addressing the patho-

physiologic mechanisms and changes in bone turnover and

bone density, there is paucity of the data addressing the

incidence of skeletal fractures 2–4 years after bariatric

surgery. In one retrospective study, a telephone survey was

obtained in 167 subjects, 12–16 months after RYGB for

morbid obesity, to evaluate the incidence of fracture [27].

The mean age of the studied subjects was 47 years with

women comprising 80 % of the participants. Six percent of

the subjects self-reported a decrease in height and 5 %

admitted appendicular skeletal fractures. Moreover, one

significant finding of the study was the high incidence of

repeated falls following surgery.

Several prospective studies have demonstrated changes

in bone mineral density (BMD) and bone turnover fol-

lowing various bariatric procedures. One prospective study

of 42 obese women (mean 37.7 years of age) 12 months

post-RYGB procedures showed a significant reduction in

the lumbar BMD and total femoral BMD of 7.4 and

10.5 %, respectively, despite calcium intake of

640–1,000 mg and vitamin D of 400–800 U/day [28].

Another study compared bone mineral density to calci-

tropic hormone in 11 non-obese, 12 obese, and 16 obese

3 years following RYGB surgery. In only subjects fol-

lowing RYGB, a significant reduction in BMD was found

at all skeletal sites. However, serum parathyroid (PTH) and

25-hydroxyvitamin D (25-OHD) levels remained unchan-

ged among all three groups while urinary deoxypyridino-

line (DPD) increased only in those following RYGB [29].

In a 1-year prospective study of 223 males and females

1 year following RYGB, ranging 21–64 years of age,

despite adequate calcium and vitamin D supplementation,

there was a significant reduction in the BMD at the femoral

neck of 9.2 %. The decrease in BMD at this site was

strongly associated with the degree of weight loss and

serum PTH. Moreover, urinary N-telopeptide (NTX)

increased significantly 3 months after surgery and

remained elevated at 12 months [17].

Four other studies collectively including 124 subjects

following RYGB showed significant fall in BMD at the

lumbar spine and hip [18, 30–32]. A very recent study of

14 women 1 year post-RYGB found that BMD did not

change at the spine or forearm, but fell significantly at the

femoral neck and total hip by 4.5 and 5.2 %, respectively.

These losses occurred despite adequate supplementation

with vitamin D and calcium [20]. This study, using high-

resolution peripheral quantitative computed tomography

(HR-pOCT), showed preferential fall in the cortical area,

cortical thickness, and total area. The changes in cortical

bone parameters were highly associated with a rise in PTH

levels. The result of this study supports that RYGB in

particular affects bone loss specifically at weight-bearing

sites including the femoral neck and total hip [17–19, 28]

(Table 1).

Bone Changes Post-gastric Banding

and Biliopancreatic Diversions

A limited number of studies have addressed the changes in

BMD, calcitropic hormones, and bone turnover markers

following gastric banding procedures and BPD. In one

study of 16 morbidly obese subjects who underwent ver-

tical banded gastroplasty (VGB), BMD, bone turnover and

calcitropic hormones were assessed 12 months after sur-

gery [33]. The results showed that BMD at the femoral

neck, trochanter, and Ward’s triangle decreased signifi-

cantly. These changes were associated with significant

increases in urinary DPD, suggestive of increased bone

turnover; however, no significant change was detected in

serum PTH. In another study, 18 patients were observed

before and after VGB. Two years following the surgery,

there was no change in BMD at the spine, but a significant

fall in BMD at the trochanter and Ward’s triangle was

noted. These changes were associated with the increased

bone resorption marker DPD; however, there was no

change in serum PTH or 25-OHD [34].

One study followed 33 subjects who underwent BPD for

10 years [35]. The results of this study showed no change

in BMD at the hip, but noted significant decrease in BMD

at the lumbar spine despite adequate supplementation with

calcium and vitamin D. These changes were associated

with a significant fall in serum 25-OHD and increases in

serum PTH, bone-specific alkaline phosphatase, and oste-

ocalcin. In another study of 63 patients undergoing BPD,

increased PTH and osteocalcin (a marker of bone turnover)

and C-terminal telopeptide of type I collagen (CTX) were

associated with decreased BMD at the spine, but not at the

hip. In another study, 96 morbidly obese patients following

BPD demonstrated an increased reduction in the bone

resorption marker, DPD, and serum PTH 2 years after the

surgery [36]. However, in this instance, BMD was not

assessed.

Pathophysiologic Mechanisms of Bone Loss Following

Bariatric Surgery

Pathophysiologic mechanisms of bone loss after bariatric

surgery are complex and may include alterations in

Clinic Rev Bone Miner Metab

123

mechanical effects on bone, nutritional deficiencies,

abnormalities in calcitropic hormone metabolism, and

alterations in energy metabolism with modulation in adi-

pokines and intestinal hormones affected following bari-

atric surgery (Fig. 1). The PubMed Review using key

words ‘‘bariatric surgery, weight loss, bone loss, and bone

metabolism’’ until 2006 has shown that bone loss fre-

quently occurs after bariatric surgery, more specifically

after RYGB procedures.

Role of Mechanical Unloading on a Skeleton

Under normal physiological circumstances, mechanical

loading has been shown to play a major role in attaining

bone mass, bone strength, and bone size [37, 38]. In

addition to bariatric surgery, several clinical conditions

associated with immobility and unloading of the skeleton

have been reported to be associated with weight loss [39–

42]. Bariatric surgery in particular has been associated with

bone loss at weight-bearing skeletal sites including the total

hip and femoral neck [17–19, 28].

The molecular mechanism of skeletal unloading has

been attributed to the up-regulation of mRNA for the

sclerostin gene (SOST). A study in SOST-deficient mice

shows that sclerostin plays an important role following

unloading the skeleton [43]. Mechanical unloading has

been demonstrated to reduce osteoblastic bone function

and consequently diminish bone formation. In this popu-

lation, increased sclerostin production through negative

regulation of WnT signaling influences osteoblastic cell

differentiation and function [44].

Role of Nutritional Deficiencies on a Skeleton

Generally, obese individuals may suffer from 25-OHD

deficiency prior to bariatric surgery [45]. The perturbation

of vitamin D metabolism may be physiological due to the

redistribution of vitamin D into the fat tissue and in part

may be acquired by way of lifestyle including insufficient

exposure to sun and social limitations [46–49]. Further-

more, in malabsorptive surgeries including RYGB, mal-

absorption of nutrients such as minerals and vitamin D due

to the loss of intestinal surface area is common [19, 28].

Therefore, as a consequence of these nutritional deficien-

cies, secondary parathyroid hormone (PTH) stimulation

will ensue and stimulate bone loss [17, 31, 50].

Restrictive bariatric surgery with the limitation of food

intake may also have adverse effects on the skeleton. The

changes in skeletal homeostasis in this population may be

associated without alteration in serum calcium and calci-

tropic hormones [34, 51].

Table 1 Bone mineral density changes following bariatric surgery

Author Number of

patients

Mean

BMI

Type of

surgery

Duration of

study (months)

Supplements Outcome BMD

Carrasco 42 45 RYGB 12 Calcium (640–1,000 mg/day)

Vitamin D (400–800 U/day)

Spine (-7.4 %)*

Total hip (-10.5 %)*

Pereira 16 33 RYGB 12 Calcium (250 mg/day)

Vitamin D (400 U/day)

Spine (-6.2 %)*

Forearm (-5.1 %)*

Femoral neck (-10.2 %)*

Fleischer 23 47 RYGB 12 Calcium (1,318 mg/day)

Vitamin D (658 U/day)

Femoral neck (-9.2 %)*

Coates 25 31 RYGB 11 Calcium (1,200 mg/day)

Vitamin D (400–800 U/day)

Spine (-3.3 %)*

Femoral neck (-5.1 %)*

Total hip (-7.8 %)*

Johnson 226 50 RYGB 36 Calcium (1,200 mg/day)

3 multivitamins

Spine (-4.5 %)*

Radius (-1.8 %)*

Total hip (-9.2 %)*

Stein 14 44 RYGB 12 Calcium (1,500–1,800 mg/day)

Vitamin D (400–800 U/day)

Femoral neck (-4.5 %)*

Total hip (-5.2 %)*

Cundy 18 43 VGB 24 Calcifediol (75 mcg/day) Ward’s triangle (-3.9 %)*

Trochanter (-4.8 %)*

Guney 16 46 VGB 12 None Femoral neck (-4.8 %)*

RYGB Roux-en-Y gastric bypass, VGB vertical banded gastroplasty

* Significant reduction in bone mineral density (BMD)

Clinic Rev Bone Miner Metab

123

Fig. 1 Pathophysiologic

mechanisms of bone loss and

kidney stone formation

following bariatric surgery

Clinic Rev Bone Miner Metab

123

Role of Calcitropic Hormone Metabolism

Abnormalities on a Skeleton

A large body of data supports the development of sec-

ondary PTH in the pathogenesis of bone loss after RYGB

and biliopancreatic diversions [31, 52, 53]. The principal

cause of bone loss after RYGB procedures has been

attributed to defective calcium absorption associated with

secondary PTH stimulation from lowered intestinal cal-

cium absorption [54, 55]. The impaired intestinal calcium

absorption has been attributed to limiting the exposure of

nutrients, which impairs the solubility of calcium salt and

fast intestinal transit [52, 53].

Vitamin D deficiency has been reported following both

malabsorptive and restrictive bariatric surgeries. Several

studies have reported low serum 25-OHD following gastric

surgery, thereby showing that vitamin D malabsorption and

secondary PTH stimulation persist despite restoration of

the calcium and vitamin D intake [16, 17, 56–58]. A ret-

rospective study showed a 73 % incidence of vitamin D

deficiency following biliopancreatic diversion procedures

[59]. Furthermore, in a study with patients following sleeve

gastrectomy, vitamin D deficiency was found in 39 % of

the subjects after 1 year despite daily multivitamin sup-

plementation [60]. Regardless of the type of bariatric sur-

gery, the prevalence of secondary PTH, increased bone

turnover, and bone mineral density loss were high in all

these populations [17, 52, 61].

Role of Energy Metabolism Alterations on a Skeleton:

Leptin

Recent advances in neurohormonal regulation of bone

metabolism have opened the door to define the significant

role of adipokines in the regulation of bone metabolism

after bariatric surgery [62]. Our knowledge of the role of

adipokines in human bone disease has not yet been fully

explored. However, leptin, which is released by adipocytes,

and its level have been shown to be significantly lower in

subjects following gastric bypass surgery compared to

obese controls [18, 63]. Nevertheless, its association with

bone mineral density has not been shown to be strong,

reflecting its dual role on bone remodeling [64]. The effect

of leptin on bone remodeling is exerted through two

independent central nervous system pathways. One mech-

anism involves the activation of sympathetic nervous sys-

tem and the stimulation of bone formation [65, 66]. The

second pathway is through the activation of cocaine–

amphetamine-regulated transcript (CART), which in turn

leads to the stimulation of receptor activator of nuclear

factor-kappa B ligand (RANK-L) expression in osteoblast,

which finally leads to enhanced bone resorption [67].

Following bariatric surgery, a fall of leptin levels is

associated with an imbalance in bone turnover, with bone

resorption exceeding bone formation thereby resulting in

net bone loss [63, 68].

Role of Energy Metabolism Alterations on a Skeleton:

Adiponectin

Adiponectin originates from adipocytes, and it has been

shown that its level is significantly increased after gastric

bypass surgery [28]. Its level has been shown to be

inversely related to the fall of bone mineral density fol-

lowing gastric bypass surgery. Its invitro effect has been

shown to be through the stimulation of RANK-L and

inhibits osteoprotegerin (OPG) in human osteoblasts [69].

Role of Energy Metabolism Alterations on a Skeleton:

Peptides

Ghrelin is a peptide produced by the stomach, which

stimulates hunger in humans [70]. Ghrelin levels diminish

following RYGB and gastric sleeve procedures. Thus, it

appears that stomach fundus plays a key role in the

secretion of this peptide [71]. The effect of the changes in

ghrelin on bone metabolism has not yet been explored in

human subjects. However, it has been shown that ghrelin

increases perforation and differentiation of osteoblasts

[72].

Peptide YY (PYY) is in the polypeptide family that

includes neuropeptide Y and pancreatic polypeptides. It is

produced by the small intestinal and colonic L cells

postprandially [70]. Following RYGB, biliopancreatic

diversions, and adjustable gastric banding, its level

increases [73–75]. At the present time, no clinical study has

shown a relationship between this peptide and bone density

in human subjects following bariatric surgery.

Glucagon-like peptides (GLP-1 and GLP-2) are also

secreted postprandially by intestinal L cells [76]. Their

levels increase significantly following RYGB procedures.

Its effect on bone turnover and bone density has not yet

been fully demonstrated; however, administering GLP-1

has been shown to improve bone density [77].

Role of Energy Metabolism Alterations on a Skeleton:

Sex Hormones

Adipocytes are the site of conversion of testosterone to

estradiol under the influence of aromatase [78]. There is

scanty literature with regard to changes in estrogen levels

following bariatric surgery. However, it has been shown

that estrogen levels generally decrease with weight loss in

both genders. Thereby, a fall in estrogen levels may either

directly or indirectly affect bone metabolism through

alteration in calcitropic hormone metabolism [79, 80].

Clinic Rev Bone Miner Metab

123

Prevalence of Kidney Stone Formation Following

Bariatric Surgery

Kidney stones have emerged as one of the main compli-

cations of modern bariatric surgeries. There are a limited

number of population-based studies concerning the preva-

lence of kidney stones following bariatric surgery. Using a

database from a national private insurance claim, one study

compared 4,690 patients who underwent RYGB with a

control group of obese subjects. It was shown that 7.5 % of

RYGB patients were diagnosed with kidney stones com-

pared with only 4.63 % of obese control patients [21].

Another retrospective study of 972 patients who underwent

RYGB found that 8.8 % admitted to kidney stones prior to

surgery and 3.2 % developed new stones postoperatively.

Of the known kidney stone formers in this population,

31 % had recurrent kidney stones following surgery.

Therefore, the stone prevalence was reported to be

approximately 70 % in the bariatric surgery population

[81].

Gastric banding was shown not to be associated with

kidney stone formation utilizing a national private insur-

ance claims database between 2002 and 2006. After gastric

banding, 1.49 % of subjects formed stones compared to

5.97 % in obese controls [82].

Pathophysiologic Mechanisms of Kidney Stone

Formation Following Bariatric Surgery

Kidney Stone Formation Following RYGB Surgery

Pathophysiologic mechanisms for kidney stone develop-

ment following RYGB surgery are numerous and include

low urine volume, high urinary oxalate, low urinary pH,

and low urinary citrate [22–26, 83–85] (Table 2).

Role of Low Urine Volume

Low urine volume is commonly encountered after RYGB

surgery as a result of low fluid intake due to a restricted

reservoir [25, 26, 83].

Role of High Urinary Oxalate

Hyperoxaluria is the most common risk factor in the

development of kidney stones in patients following RYBG

surgery. In fact, it has been detected in one- to two-thirds of

patients in this population [22–26, 83–85]. This abnor-

mality may occur early or late depending on the type of

surgery, health of the patient, and differences in dietary

intake of protein, fat, calcium, and oxalate [26, 83, 85].

Despite the common belief that RYGB surgery overcame

the complication of kidney stone formation previously

caused by Jegunoileal (JI) [86], a 2005 study showed an

association between RYGB and kidney stone formation

[22]. In this study, 21 out of 23 patients who underwent

RYGB developed kidney stones associated with highly

Table 2 Kidney stone risk profiles following RYGB surgery

Before RYGB Following RYGB

Urinary oxalate

Nelson et al. [22] N/A 79

Sinha et al. [23] 31 ± 16 65 ± 39*

Asplin and Coe [24] N/A 85 ± 44*

Duffey et al. [26] 31 ± 10 41 ± 18*

Penniston et al. [97] N/A 48 ± 4

Park et al. [25] 32 (median) 40 (median)*

Maalouf et al. [93] N/A 45 ± 21*

Patel et al. [84] N/A 61 ± 4*

Kumar et al. [85] 26 ± 13 32 ± 11 (NS)

Froeder et al. [94] N/A 26 (median)(NS)

Urinary Citrate (mg/day)

Sinha et al. [23] 660 ± 297 444 ± 376(NS)

Asplin and Coe [24] N/A 477 ± 330*

Penniston et al. [97] N/A 441 ± 71*

Park et al. [25] 675 (median) 456 (median)*

Maalouf et al. [93] N/A 358 ± 357*

Patel et al. [84] N/A 621 ± 40*

Froeder et al. [94] N/A 472 (median)(NS)

Urinary pH

Asplin and Coe [24] N/A 5.72 ± 0.31*

Sinha et al. [23] 5.96 ± 0.38 5.78 ± 0.59(NS)

Duffey et al. [26] 5.82 ± 0.54 5.66 ± 0.43(NS)

Park et al. [25] 6.03 (median) 5.75 (median) (NS)

Froeder et al. [94] N/A 5.78 (median) (NS)

Urinary volume (mg/day)

Duffey et al. [26] 1,380 ± 400 900 ± 430*

Park et al. [25] 1,800 (median) 1,440*

Maalouf et al. [93] N/A 1,900 ± 900 (NS)

Kumar et al. [85] 2,091 ± 768 1,316 ± 540*

Froeder et al. [94] N/A 1,140*

Urinary calcium (mg/day)

Sinha et al. [23] 206 ± 111 112 ± 92*

Asplin and Coe [24] N/A 141 ± 61*

Duffey et al. [26] 206 ± 111 112 ± 92*

Fleischer et al. [17] 161 ± 22 92 ± 15*

Penniston et al. [97] N/A 100 ± 12*

Park et al. [25] 176 (median) 135* (median)

Maalouf et al. [93] N/A 115 ± 93*

Froeder et al. [94] N/A 89* (median)

N/A not available, NS statistically nonsignificant

* Significant compared to control

Clinic Rev Bone Miner Metab

123

elevated urinary oxalate. Moreover, 2 patients developed

acute renal injury due to high oxalate burden in the kidney.

Following that report, several investigators reported high

incidence of kidney stone, hyperoxaluria, and elevated

urinary supersaturation with respect to calcium oxalate [23,

24, 84, 85] (Fig. 1).

The underlying pathophysiologic mechanisms for

hyperoxaluria following RYGB procedure have not yet

been fully elucidated. (1) One purported mechanism has

linked hyperoxaluria to intestinal fat malabsorption [85,

87]. In this scheme, the unabsorbed fat increases free

luminal oxalate content by binding to calcium in intestinal

lumen and thereby enhancing intestinal oxalate absorption.

However, few studies have alluded to fecal fat excretion in

this population [85, 88]. (2) Moreover, it has been sug-

gested that changes in intestinal microbial flora following

bariatric surgery can potentially modify the colonization of

lower intestinal flora with respect to oxalobacter formig-

enes, which is recognized for its capacity to degrade oxa-

late [89, 90]. However, this pathogenetic scheme has not

yet been well elucidated among the population. (3) Yet,

another mechanism has been suggested to be due to

increased permeability of the colon as a result of exposure

to unconjugated bile acids and long-chain fatty acids fol-

lowing RYGB procedures [91, 92].

Role of Low Urinary pH

In two studies, low urinary pH has been shown following

RYGB procedure [25, 93], associated with supersaturation

of uric acid.

Role of Low Urinary Citrate

Low urinary citrate is found in the majority of, but not all,

patients following RYGB [23, 24, 26, 84, 85]. However, its

prevalence has been demonstrated to vary with different

reports ranging between 34 and 63 % [25, 83, 93, 94].

Hypocitraturia occurs in this population in the absence of

overt metabolic acidosis, suggesting the homeostatic role

of the skeleton in the buffering of an excessive acid load

[95].

Kidney Stone Formation Following Restrictive Surgery

Two studies explore the biochemical profile following

gastric banding and/or sleeve gastrectomy. In one study, a

total of 18 patients (14 with gastric banding and 4 with stiff

gastrectomy) were studied for 6 months following surgery.

The urinary kidney stone risk profiles were compared with

the RYGB cohort [96]. In this study, urinary oxalate

excretion and urinary supersaturation calcium oxalate were

comparable to control subjects, but significantly lower in

restrictive cohorts (gastric banding and sleeve gastrectomy)

compared with the RYGB population. Similar findings

were obtained in another cohort comparing 21 subjects

who underwent RYGB and 12 who underwent gastric

banding. The results showed that patients with gastric

banding had low urinary volume, but did not have other

kidney stone risk factors that were detected in the cohort

who underwent RYGB, including hypocitraturia and

hyperoxaluria [97].

Role of Urinary Calcium

Low urinary calcium is commonly encountered following

RYGB procedures, which plays an inhibitory role against

calcium oxalate crystallization by overriding the effect of

hypocitraturia and hyperoxaluria [17, 23, 25, 26, 93, 97].

The underlying mechanism of hypocalciuria is associated

with impaired intestinal calcium absorption [54, 98].

Potential Treatment Approach Toward Skeletal Bone

Disease and Kidney Stone Formation Following

Bariatric Surgery

Despite the availability of hard evidence of bone fracture

efficacy data, the American Association of Clinical Endo-

crinologists (AACE), the Obesity Society (TOS), and the

American Society of Metabolic and Bariatric Surgery

(ASMBS) have recommended that calcium supplementa-

tion be considered after bariatric surgery [99]. These

guidelines advocate the daily treatment 1,200–2,000 mg of

calcium and ergocalciferol at 50,000 U 1–2 times/week

and higher doses up to 50,000–150,000 U/day for severe

vitamin D deficiency. In rare instances when the subject

does not respond to the maximal does of ergocalciferol,

calcitriol treatment may be the drug of choice.

The above recommendation is pathophysiologically

logical since impaired intestinal calcium absorption occurs

[54, 98] associated with vitamin D deficiency and sec-

ondary parathyroid stimulation, which commonly befalls

this population [17, 19, 28, 31, 50]. In addition, GI alkali

loss occurs following RYGB surgery increasing the risk of

kidney stone complication [22, 23, 93].

AACE/TOS/ASMBS guidelines recommend calcium

citrate tablets as the preferred choice in this population due

to its superior intestinal bioavailability [100, 101]. The low

intestinal bioavailability of calcium carbonate was shown

to be due to diminished solubility as a result of the lack of

exposure of duodenum, the main site of calcium absorp-

tion, to gastric acid secretion and also due to rapid intes-

tinal transit time [102]. However, the latter was based on

the study in postmenopausal women comparing the tablet

formulation of calcium carbonate and calcium citrate.

Clinic Rev Bone Miner Metab

123

Since then, it was found that calcium citrate in tablet for-

mulation is not efficiently absorbed in the RYGB popula-

tion [98, 103]. Moreover, both calcium citrate and calcium

carbonate in tablet forms were not shown to ameliorate

PTH secretion in RYGB patients [98, 100, 103].

It is imperative to use a product to overcome the above

limitations of calcium citrate tablets and reverse the path-

ophysiologic derangements responsible for the develop-

ment of bone loss and kidney stone formation. Recently, in

a short-term, randomized, crossover placebo control trial

study in 24 patients following RYGB, it was shown that

effervescent potassium calcium citrate (PCC) significantly

inhibited bone turnover by lowering PTH secretion [104]

(Fig. 2a, b). Moreover, alkali provision from PCC

increased urinary pH and citrate, while attenuating a rise in

urinary calcium from increased intestinal calcium absorp-

tion. The aforementioned changes were associated with an

increased inhibition against calcium oxalate agglomera-

tions and reducing urinary supersaturation of uric acid

(Fig. 3a, b). Despite the availability of hard evidence such

as bone fracture efficacy and kidney stone incidence, this

novel approach appears promising and requires further

investigation.

Although bisphosphonates are the drug of choice in the

management of senile osteoporosis, there is concern with

the use of this drug in the RYGB population. This stems

from case reports suggesting that underlying intestinal

calcium and vitamin D malabsorption may accentuate the

development of severe hypocalcemia after the administra-

tion of bisphosphonate in this population [105].

Conclusion

The benefits of bariatric surgery in sustaining weight loss

and its increased benefit in the reduction in cardiovascular

and diabetic complications have been well established.

However, metabolic complications including bone loss and

nephrolithiasis are almost inevitable. It has been shown that

RYGB is the most effective among all procedures. How-

ever, it carries the highest metabolic risk. Therefore, it is

imperative that patients receive prophylactic measures to

avoid this complication. To date, the use of commonly

prescribed over-the-counter calcium supplement and vita-

min D supplementation has not been shown to avert bone

loss. This limitation has been to a major extent due to

significantly impaired intestinal calcium absorption.

Despite treatment with calcium supplements in tablet

Fig. 2 [104]: a Effect of PCC and placebo on serum calcium (Ca)

and PTH. Vertical bars indicate mean ± SD. Dashed horizontal line

represents upper normal limit of serum PTH. b Effect of PCC and

placebo on serum CTX and urinary deoxypyridinoline (DPD). Dashed

horizontal lines represent upper normal limits. **P \ .01; �P \ .001

between 2 phasesFig. 3 [104]: a Effect of PCC and placebo on Supersaturation Index

(SI) of calcium oxalate (Ca oxalate), brushite, and uric acid. Dashed

horizontal line indicates saturation value. b Effect of PCC and

placebo on agglomeration inhibition of calcium oxalate. Data

individually depicted for 8 urine samples, in which concentrations

of citrate, calcium, and pH were altered to mimic those of PCC and

placebo phases

Clinic Rev Bone Miner Metab

123

formulation, it appears that a pre-solubilized calcium cit-

rate and potassium citrate formulation will overcome these

limitations and reverse underlying pathophysiologic

abnormalities responsible for the development of both bone

loss and kidney stone formation. A preliminary study has

suggested an improvement in bone turnover and reduction

in kidney stone risk in this population. However, despite

this evidence, there is a lack of hard data showing the

effectiveness of this formulation in the reduction in fracture

and kidney stone incidence. Future investigation is needed

to explore these effects.

Acknowledgments The author would like to acknowledge Ashlei

L. Johnson for her role in the editorial process of this manuscript.

Disclosures

Conflict of interest Khashayar Sakhaee declares that he has no

conflict of interest.

Animal/Human Studies This article does not contain any studies

with human or animal subjects performed by the author.

References

1. Adams TD, Gress RE, Smith SC, Halverson RC, Simper SC,

Rosamond WD, et al. Long-term mortality after gastric bypass

surgery. N Engl J Med. 2007;357(8):753–61. doi:10.1056/

NEJMoa066603.

2. Brolin RE. Bariatric surgery and long-term control of morbid

obesity. JAMA. 2002;288(22):2793–6.

3. Sjostrom L, Narbro K, Sjostrom CD, Karason K, Larsson B,

Wedel H, et al. Effects of bariatric surgery on mortality in

Swedish obese subjects. N Engl J Med. 2007;357(8):741–52.

doi:10.1056/NEJMoa066254.

4. Flegal KM, Carroll MD, Ogden CL, Johnson CL. Prevalence

and trends in obesity among US adults, 1999–2000. JAMA.

2002;288(14):1723–7.

5. Yanovski SZ, Yanovski JA. Obesity. N Engl J Med.

2002;346(8):591–602. doi:10.1056/NEJMra012586.

6. Sjostrom L, Lindroos AK, Peltonen M, Torgerson J, Bouchard

C, Carlsson B, et al. Lifestyle, diabetes, and cardiovascular risk

factors 10 years after bariatric surgery. N Engl J Med.

2004;351(26):2683–93. doi:10.1056/NEJMoa035622.

7. Balsiger BM, Murr MM, Poggio JL, Sarr MG. Bariatric surgery.

Surgery for weight control in patients with morbid obesity. Med

Clin North Am. 2000;84(2):477–89.

8. Halverson JD, Wise L, Wazna MF, Ballinger WF. Jejunoileal

bypass for morbid obesity. A critical appraisal. Am J Med.

1978;64(3):461–75.

9. Scopinaro N, Gianetta E, Adami GF, Friedman D, Traverso E,

Marinari GM, et al. Biliopancreatic diversion for obesity at

eighteen years. Surgery. 1996;119(3):261–8.

10. Scopinaro N, Marinari GM, Camerini G. Laparoscopic standard

biliopancreatic diversion: technique and preliminary results. Obes

Surg. 2002;12(2):241–4. doi:10.1381/096089202762552692.

11. Deitel M, Shahi B, Anand PK, Deitel FH, Cardinell DL. Long-

term outcome in a series of jejunoileal bypass patients. Obes

Surg. 1993;3(3):247–52. doi:10.1381/096089293765559250.

12. Steinbrook R. Surgery for severe obesity. N Engl J Med.

2004;350(11):1075–9. doi:10.1056/NEJMp048029.

13. Schauer PR, Kashyap SR, Wolski K, Brethauer SA, Kirwan JP,

Pothier CE, et al. Bariatric surgery versus intensive medical

therapy in obese patients with diabetes. N Engl J Med.

2012;366(17):1567–76. doi:10.1056/NEJMoa1200225.

14. Mingrone G, Panunzi S, De Gaetano A, Guidone C, Iaconelli A,

Leccesi L, et al. Bariatric surgery versus conventional medical

therapy for type 2 diabetes. N Engl J Med. 2012;366(17):1577–85.

doi:10.1056/NEJMoa1200111.

15. Mahdy T, Atia S, Farid M, Adulatif A. Effect of Roux-en Y gastric

bypass on bone metabolism in patients with morbid obesity:

Mansoura experiences. Obes Surg. 2008;18(12):1526–31. doi:10.

1007/s11695-008-9653-1.

16. De Prisco C, Levine SN. Metabolic bone disease after gastric

bypass surgery for obesity. Am J Med Sci. 2005;329(2):57–61.

17. Fleischer J, Stein EM, Bessler M, Della Badia M, Restuccia N,

Olivero-Rivera L, et al. The decline in hip bone density after

gastric bypass surgery is associated with extent of weight loss.

J Clin Endocrinol Metab. 2008;93(10):3735–40. doi:10.1210/jc.

2008-0481.

18. Coates PS, Fernstrom JD, Fernstrom MH, Schauer PR, Green-

span SL. Gastric bypass surgery for morbid obesity leads to an

increase in bone turnover and a decrease in bone mass. J Clin

Endocrinol Metab. 2004;89(3):1061–5.

19. Vilarrasa N, San Jose P, Garcia I, Gomez-Vaquero C, Miras PM,

de Gordejuela AG, et al. Evaluation of bone mineral density loss in

morbidly obese women after gastric bypass: 3-year follow-up.

Obes Surg. 2011;21(4):465–72. doi:10.1007/s11695-010-0338-1.

20. Stein EM, Carrelli A, Young P, Bucovsky M, Zhang C, Schrope

B, et al. Bariatric surgery results in cortical bone loss. J Clin

Endocrinol Metab. 2013;98(2):541–9. doi:10.1210/jc.2012-

2394.

21. Matlaga BR, Shore AD, Magnuson T, Clark JM, Johns R,

Makary MA. Effect of gastric bypass surgery on kidney stone

disease. J Urol. 2009;181(6):2573–7. doi:10.1016/j.juro.2009.

02.029.

22. Nelson WK, Houghton SG, Milliner DS, Lieske JC, Sarr MG.

Enteric hyperoxaluria, nephrolithiasis, and oxalate nephropathy:

potentially serious and unappreciated complications of Roux-en-

Y gastric bypass. Surg Obes Relat Dis. 2005;1(5):481–5. doi:10.

1016/j.soard.2005.07.002.

23. Sinha MK, Collazo-Clavell ML, Rule A, Milliner DS, Nelson

W, Sarr MG, et al. Hyperoxaluric nephrolithiasis is a compli-

cation of Roux-en-Y gastric bypass surgery. Kidney Int.

2007;72(1):100–7. doi:10.1038/sj.ki.5002194.

24. Asplin JR, Coe FL. Hyperoxaluria in kidney stone formers treated

with modern bariatric surgery. J Urol. 2007;177(2):565–9. doi:10.

1016/j.juro.2006.09.033.

25. Park AM, Storm DW, Fulmer BR, Still CD, Wood GC, Hartle JE

2nd. A prospective study of risk factors for nephrolithiasis after

Roux-en-Y gastric bypass surgery. J Urol. 2009;182(5):2334–9.

doi:10.1016/j.juro.2009.07.044.

26. Duffey BG, Pedro RN, Makhlouf A, Kriedberg C, Stessman M,

Hinck B, et al. Roux-en-Y gastric bypass is associated with early

increased risk factors for development of calcium oxalate

nephrolithiasis. J Am Coll Surg. 2008;206(3):1145–53. doi:10.

1016/j.jamcollsurg.2008.01.015.

27. Berarducci A, Haines K, Murr MM. Incidence of bone loss,

falls, and fractures after Roux-en-Y gastric bypass for morbid

obesity. Appl Nurs Res. 2009;22(1):35–41. doi:10.1016/j.apnr.

2007.03.004.

28. Carrasco F, Ruz M, Rojas P, Csendes A, Rebolledo A, Codoceo J,

et al. Changes in bone mineral density, body composition and

adiponectin levels in morbidly obese patients after bariatric surgery.

Obes Surg. 2009;19(1):41–6. doi:10.1007/s11695-008-9638-0.

29. Pereira FA, de Castro JA, dos Santos JE, Foss MC, Paula FJ.

Impact of marked weight loss induced by bariatric surgery on

Clinic Rev Bone Miner Metab

123

bone mineral density and remodeling. Braz J Med Biol Res.

2007;40(4):509–17.

30. von Mach MA, Stoeckli R, Bilz S, Kraenzlin M, Langer I, Keller

U. Changes in bone mineral content after surgical treatment of

morbid obesity. Metabolism. 2004;53(7):918–21.

31. Goode LR, Brolin RE, Chowdhury HA, Shapses SA. Bone and

gastric bypass surgery: effects of dietary calcium and vitamin D.

Obes Res. 2004;12(1):40–7. doi:10.1038/oby.2004.7.

32. Bano G, Rodin DA, Pazianas M, Nussey SS. Reduced bone

mineral density after surgical treatment for obesity. Int J Obes

Relat Metab Disord. 1999;23(4):361–5.

33. Guney E, Kisakol G, Ozgen G, Yilmaz C, Yilmaz R, Kabalak T.

Effect of weight loss on bone metabolism: comparison of ver-

tical banded gastroplasty and medical intervention. Obes Surg.

2003;13(3):383–8. doi:10.1381/096089203765887705.

34. Cundy T, Evans MC, Kay RG, Dowman M, Wattie D, Reid IR.

Effects of vertical-banded gastroplasty on bone and mineral

metabolism in obese patients. Br J Surg. 1996;83(10):1468–72.

35. Marceau P, Biron S, Lebel S, Marceau S, Hould FS, Simard S,

et al. Does bone change after biliopancreatic diversion? J Gas-

trointest Surg. 2002;6(5):690–8.

36. Moreiro J, Ruiz O, Perez G, Salinas R, Urgeles JR, Riesco M, et al.

Parathyroid hormone and bone marker levels in patients with

morbid obesity before and after biliopancreatic diversion. Obes

Surg. 2007;17(3):348–54. doi:10.1007/s11695-007-9063-9.

37. Felson DT, Zhang Y, Hannan MT, Anderson JJ. Effects of

weight and body mass index on bone mineral density in men and

women: the Framingham study. J Bone Miner Res. 1993;

8(5):567–73. doi:10.1002/jbmr.5650080507.

38. Schoenau E, Frost HM. The ‘‘muscle-bone unit’’ in children and

adolescents. Calcif Tissue Int. 2002;70(5):405–7. doi:10.1007/

s00223-001-0048-8.

39. Maimoun L, Fattal C, Micallef JP, Peruchon E, Rabischong P.

Bone loss in spinal cord-injured patients: from physiopathology

to therapy. Spinal Cord. 2006;44(4):203–10. doi:10.1038/sj.sc.

3101832.

40. Zerwekh JE, Ruml LA, Gottschalk F, Pak CY. The effects of

twelve weeks of bed rest on bone histology, biochemical

markers of bone turnover, and calcium homeostasis in eleven

normal subjects. J Bone Miner Res. 1998;13(10):1594–601.

doi:10.1359/jbmr.1998.13.10.1594.

41. Suva LJ. Sclerostin and the unloading of bone. J Bone Miner

Res. 2009;24(10):1649–50. doi:10.1359/jbmr.090815.

42. Schneider V, Oganov V, LeBlanc A, Rakmonov A, Taggart L,

Bakulin A, et al. Bone and body mass changes during space

flight. Acta Astronaut. 1995;36(8–12):463–6.

43. Lin C, Jiang X, Dai Z, Guo X, Weng T, Wang J, et al. Sclerostin

mediates bone response to mechanical unloading through

antagonizing Wnt/beta-catenin signaling. J Bone Miner Res.

2009;24(10):1651–61. doi:10.1359/jbmr.090411.

44. Day TF, Guo X, Garrett-Beal L, Yang Y. Wnt/beta-catenin

signaling in mesenchymal progenitors controls osteoblast and

chondrocyte differentiation during vertebrate skeletogenesis.

Dev Cell. 2005;8(5):739–50. doi:10.1016/j.devcel.2005.03.016.

45. Compher CW, Badellino KO, Boullata JI. Vitamin D and the

bariatric surgical patient: a review. Obes Surg. 2008;

18(2):220–4. doi:10.1007/s11695-007-9289-6.

46. Earthman CP, Beckman LM, Masodkar K, Sibley SD. The link

between obesity and low circulating 25-hydroxyvitamin D

concentrations: considerations and implications. Int J Obes.

2012;36(3):387–96. doi:10.1038/ijo.2011.119.

47. Wortsman J, Matsuoka LY, Chen TC, Lu Z, Holick MF.

Decreased bioavailability of vitamin D in obesity. Am J Clin

Nutr. 2000;72(3):690–3.

48. Grethen E, McClintock R, Gupta CE, Jones R, Cacucci BM,

Diaz D, et al. Vitamin D and hyperparathyroidism in obesity.

J Clin Endocrinol Metab. 2011;96(5):1320–6. doi:10.1210/jc.

2010-2202.

49. Stein EM, Strain G, Sinha N, Ortiz D, Pomp A, Dakin G, et al.

Vitamin D insufficiency prior to bariatric surgery: risk factors and

a pilot treatment study. Clin Endocrinol. 2009;71(2):176–83.

doi:10.1111/j.1365-2265.2008.03470.x.

50. Avgerinos DV, Leitman IM, Martinez RE, Liao EP. Evaluation

of markers for calcium homeostasis in a population of obese

adults undergoing gastric bypass operations. J Am Coll Surg.

2007;205(2):294–7. doi:10.1016/j.jamcollsurg.2007.02.078.

51. Pugnale N, Giusti V, Suter M, Zysset E, Heraief E, Gaillard RC,

et al. Bone metabolism and risk of secondary hyperparathyroid-

ism 12 months after gastric banding in obese pre-menopausal

women. Int J Obes Relat Metab Disord. 2003;27(1):110–6.

doi:10.1038/sj.ijo.0802177.

52. Youssef Y, Richards WO, Sekhar N, Kaiser J, Spagnoli A,

Abumrad N, et al. Risk of secondary hyperparathyroidism after

laparoscopic gastric bypass surgery in obese women. Surg En-

dosc. 2007;21(8):1393–6. doi:10.1007/s00464-007-9228-6.

53. Valderas JP, Velasco S, Solari S, Liberona Y, Viviani P, Maiz

A, et al. Increase of bone resorption and the parathyroid hor-

mone in postmenopausal women in the long-term after Roux-en-

Y gastric bypass. Obes Surg. 2009;19(8):1132–8. doi:10.1007/

s11695-009-9890-y.

54. Riedt CS, Brolin RE, Sherrell RM, Field MP, Shapses SA. True

fractional calcium absorption is decreased after Roux-en-Y

gastric bypass surgery. Obesity. 2006;14(11):1940–8. doi:10.

1038/oby.2006.226.

55. Williams SE. Metabolic bone disease in the bariatric surgery

patient. J Obes. 2011;2011:634614. doi:10.1155/2011/634614.

56. Johnson JM, Maher JW, DeMaria EJ, Downs RW, Wolfe LG,

Kellum JM. The long-term effects of gastric bypass on vitamin

D metabolism. Ann Surg. 2006;243(5):701–4; discussion 4–5.

doi:10.1097/01.sla.0000216773.47825.c1.

57. El-Kadre LJ, Rocha PR, de Almeida Tinoco ac, Tinoco Rc. Calcium

metabolism in pre- and postmenopausal morbidly obese women at

baseline and after laparoscopic Roux-en-Y gastric bypass. Obes

Surg. 2004;14(8):1062–6. doi:10.1381/0960892041975505.

58. Madan AK, Orth WS, Tichansky DS, Ternovits CA. Vitamin

and trace mineral levels after laparoscopic gastric bypass. Obes

Surg. 2006;16(5):603–6. doi:10.1381/096089206776945057.

59. Khandalavala BN, Hibma PP, Fang X. Prevalence and persis-

tence of vitamin D deficiency in biliopancreatic diversion

patients: a retrospective study. Obes Surg. 2010;20(7):881–4.

doi:10.1007/s11695-010-0185-0.

60. Aarts EO, Janssen IM, Berends FJ. The gastric sleeve: losing

weight as fast as micronutrients? Obes Surg. 2011;21(2):207–11.

doi:10.1007/s11695-010-0316-7.

61. Balsa JA, Botella-Carretero JI, Peromingo R, Caballero C,

Munoz-Malo T, Villafruela JJ, et al. Chronic increase of bone

turnover markers after biliopancreatic diversion is related to

secondary hyperparathyroidism and weight loss. Relation with

bone mineral density. Obes Surg. 2010;20(4):468–73. doi:10.

1007/s11695-009-0028-z.

62. Wucher H, Ciangura C, Poitou C, Czernichow S. Effects of

weight loss on bone status after bariatric surgery: association

between adipokines and bone markers. Obes Surg. 2008;

18(1):58–65. doi:10.1007/s11695-007-9258-0.

63. Garcia de la Torre N, Rubio MA, Bordiu E, Cabrerizo L,

Aparicio E, Hernandez C et al. Effects of weight loss after ba-

riatric surgery for morbid obesity on vascular endothelial growth

factor-A, adipocytokines, and insulin. J Clin Endocrinol Metab.

2008;93(11):4276–81. doi:10.1210/jc.2007-1370.

64. Thomas T, Burguera B. Is leptin the link between fat and bone

mass? J Bone Miner Res. 2002;17(9):1563–9. doi:10.1359/jbmr.

2002.17.9.1563.

Clinic Rev Bone Miner Metab

123

65. Karsenty G, Oury F. The central regulation of bone mass, the

first link between bone remodeling and energy metabolism.

J Clin Endocrinol Metab. 2010;95(11):4795–801. doi:10.1210/

jc.2010-1030.

66. Yadav VK, Oury F, Suda N, Liu ZW, Gao XB, Confavreux C,

et al. A serotonin-dependent mechanism explains the leptin

regulation of bone mass, appetite, and energy expenditure. Cell.

2009;138(5):976–89. doi:10.1016/j.cell.2009.06.051.

67. Karsenty G. Convergence between bone and energy homeostases:

leptin regulation of bone mass. Cell Metab. 2006;4(5):341–8.

doi:10.1016/j.cmet.2006.10.008.

68. Bruno C, Fulford AD, Potts JR, McClintock R, Jones R, Cacucci

BM, et al. Serum markers of bone turnover are increased at six

and 18 months after Roux-en-Y bariatric surgery: correlation

with the reduction in leptin. J Clin Endocrinol Metab.

2010;95(1):159–66. doi:10.1210/jc.2009-0265.

69. Luo XH, Guo LJ, Xie H, Yuan LQ, Wu XP, Zhou HD, et al.

Adiponectin stimulates RANKL and inhibits OPG expression in

human osteoblasts through the MAPK signaling pathway.

J Bone Miner Res. 2006;21(10):1648–56. doi:10.1359/jbmr.

060707.

70. Cummings DE, Overduin J. Gastrointestinal regulation of food

intake. J Clin Invest. 2007;117(1):13–23. doi:10.1172/JCI30227.

71. Tymitz K, Engel A, McDonough S, Hendy MP, Kerlakian G.

Changes in ghrelin levels following bariatric surgery: review of

the literature. Obes Surg. 2011;21(1):125–30. doi:10.1007/

s11695-010-0311-z.

72. Huda MS, Dovey T, Wong SP, English PJ, Halford J, McCul-

loch P, et al. Ghrelin restores ‘lean-type’ hunger and energy

expenditure profiles in morbidly obese subjects but has no effect

on postgastrectomy subjects. Int J Obes. 2009;33(3):317–25.

doi:10.1038/ijo.2008.270.

73. Garcia-Fuentes E, Garrido-Sanchez L, Garcia-Almeida JM,

Garcia-Arnes J, Gallego-Perales JL, Rivas-Marin J, et al. Dif-

ferent effect of laparoscopic Roux-en-Y gastric bypass and open

biliopancreatic diversion of Scopinaro on serum PYY and

ghrelin levels. Obes Surg. 2008;18(11):1424–9. doi:10.1007/

s11695-008-9560-5.

74. Reinehr T, Roth CL, Schernthaner GH, Kopp HP, Kriwanek S,

Schernthaner G. Peptide YY and glucagon-like peptide-1 in

morbidly obese patients before and after surgically induced

weight loss. Obes Surg. 2007;17(12):1571–7. doi:10.1007/

s11695-007-9323-8.

75. Karamanakos SN, Vagenas K, Kalfarentzos F, Alexandrides TK.

Weight loss, appetite suppression, and changes in fasting and

postprandial ghrelin and peptide-YY levels after Roux-en-Y

gastric bypass and sleeve gastrectomy: a prospective, double

blind study. Ann Surg. 2008;247(3):401–7. doi:10.1097/SLA.

0b013e318156f012.

76. Bollag RJ, Zhong Q, Phillips P, Min L, Zhong L, Cameron R,

et al. Osteoblast-derived cells express functional glucose-

dependent insulinotropic peptide receptors. Endocrinology.

2000;141(3):1228–35.

77. Nuche-Berenguer B, Moreno P, Esbrit P, Dapia S, Caeiro JR,

Cancelas J, et al. Effect of GLP-1 treatment on bone turnover in

normal, type 2 diabetic, and insulin-resistant states. Calcif Tis-

sue Int. 2009;84(6):453–61. doi:10.1007/s00223-009-9220-3.

78. Grodin JM, Siiteri PK, MacDonald PC. Source of estrogen

production in postmenopausal women. J Clin Endocrinol Metab.

1973;36(2):207–14.

79. Hammoud A, Gibson M, Hunt SC, Adams TD, Carrell DT, Ko-

lotkin RL, et al. Effect of Roux-en-Y gastric bypass surgery on

the sex steroids and quality of life in obese men. J Clin Endo-

crinol Metab. 2009;94(4):1329–32. doi:10.1210/jc.2008-1598.

80. Gallagher JC, Riggs BL, DeLuca HF. Effect of estrogen on

calcium absorption and serum vitamin D metabolites in

postmenopausal osteoporosis. J Clin Endocrinol Metab.

1980;51(6):1359–64.

81. Durrani O, Morrisroe S, Jackman S, Averch T. Analysis of stone

disease in morbidly obese patients undergoing gastric bypass

surgery. J Endourol. 2006;20(10):749–52. doi:10.1089/end.

2006.20.749.

82. Semins MJ, Matlaga BR, Shore AD, Steele K, Magnuson T,

Johns R, et al. The effect of gastric banding on kidney stone

disease. Urology. 2009;74(4):746–9. doi:10.1016/j.urology.

2009.04.093.

83. Duffey BG, Alanee S, Pedro RN, Hinck B, Kriedberg C, Ikra-

muddin S, et al. Hyperoxaluria is a long-term consequence of

Roux-en-Y Gastric bypass: a 2-year prospective longitudinal

study. J Am Coll Surg. 2010;211(1):8–15. doi:10.1016/j.

jamcollsurg.2010.03.007.

84. Patel BN, Passman CM, Fernandez A, Asplin JR, Coe FL, Kim

SC, et al. Prevalence of hyperoxaluria after bariatric surgery.

J Urol. 2009;181(1):161–6. doi:10.1016/j.juro.2008.09.028.

85. Kumar R, Lieske JC, Collazo-Clavell ML, Sarr MG, Olson ER,Vrtiska TJ, et al. Fat malabsorption and increased intestinal

oxalate absorption are common after Roux-en-Y gastric bypass

surgery. Surgery. 2011;149(5):654–61. doi:10.1016/j.surg.2010.

11.015.

86. Requarth JA, Burchard KW, Colacchio TA, Stukel TA, Mott

LA, Greenberg ER, et al. Long-term morbidity following jeju-

noileal bypass. The continuing potential need for surgical

reversal. Arch Surg. 1995;130(3):318–25.

87. Ferraz RR, Tiselius HG, Heilberg IP. Fat malabsorption induced

by gastrointestinal lipase inhibitor leads to an increase in urinary

oxalate excretion. Kidney Int. 2004;66(2):676–82. doi:10.1111/

j.1523-1755.2004.00790.x.

88. Odstrcil EA, Martinez JG, Santa Ana CA, Xue B, Schneider RE,

Steffer KJ, et al. The contribution of malabsorption to the

reduction in net energy absorption after long-limb Roux-en-Y

gastric bypass. Am J Clin Nutr. 2010;92(4):704–13. doi:10.

3945/ajcn.2010.29870.

89. Li JV, Ashrafian H, Bueter M, Kinross J, Sands C, le Roux CW,

et al. Metabolic surgery profoundly influences gut microbial-

host metabolic cross-talk. Gut. 2011;60(9):1214–23. doi:10.

1136/gut.2010.234708.

90. Kaufman DW, Kelly JP, Curhan GC, Anderson TE, Dretler SP,

Preminger GM, et al. Oxalobacter formigenes may reduce the

risk of calcium oxalate kidney stones. J Am Soc Nephrol.

2008;19(6):1197–203. doi:10.1681/ASN.2007101058.

91. Freel RW, Hatch M, Earnest DL, Goldner AM. Oxalate trans-

port across the isolated rat colon. A re-examination. Biochim

Biophys Acta. 1980;600(3):838–43.

92. Dobbins JW, Binder HJ. Effect of bile salts and fatty acids on

the colonic absorption of oxalate. Gastroenterology. 1976;

70(6):1096–100.

93. Maalouf NM, Tondapu P, Guth ES, Livingston EH, Sakhaee K.

Hypocitraturia and hyperoxaluria after Roux-en-Y gastric

bypass surgery. J Urol. 2010;183(3):1026–30. doi:10.1016/j.

juro.2009.11.022.

94. Froeder L, Arasaki CH, Malheiros CA, Baxmann AC, Heilberg

IP. Response to dietary oxalate after bariatric surgery. Clin J Am

Soc Nephrol. 2012;7(12):2033–40. doi:10.2215/CJN.02560312.

95. Alpern RJ, Sakhaee K. The clinical spectrum of chronic meta-

bolic acidosis: homeostatic mechanisms produce significant

morbidity. Am J Kidney Dis. 1997;29(2):291–302.

96. Semins MJ, Asplin JR, Steele K, Assimos DG, Lingeman JE,

Donahue S, et al. The effect of restrictive bariatric surgery on

urinary stone risk factors. Urology. 2010;76(4):826–9. doi:10.

1016/j.urology.2010.01.037.

97. Penniston KL, Kaplon DM, Gould JC, Nakada SY. Gastric band

placement for obesity is not associated with increased urinary

Clinic Rev Bone Miner Metab

123

risk of urolithiasis compared to bypass. J Urol.

2009;182(5):2340–6. doi:10.1016/j.juro.2009.07.041.

98. Tondapu P, Provost D, Adams-Huet B, Sims T, Chang C,

Sakhaee K. Comparison of the absorption of calcium carbonate

and calcium citrate after Roux-en-Y gastric bypass. Obes Surg.

2009;19(9):1256–61. doi:10.1007/s11695-009-9850-6.

99. Mechanick JI, Kushner RF, Sugerman HJ, Gonzalez-Campoy

JM, Collazo-Clavell ML, Guven S, et al. American Association

of Clinical Endocrinologists, The Obesity Society, and Ameri-

can Society for Metabolic AND Bariatric Surgery Medical

Guidelines for Clinical Practice for the perioperative nutritional,

metabolic, and nonsurgical support of the bariatric surgery

patient. Surg Obes Relat Dis. 2008;4(5 Suppl):S109–84. doi:10.

1016/j.soard.2008.08.009.

100. Heller HJ, Greer LG, Haynes SD, Poindexter JR, Pak CY.

Pharmacokinetic and pharmacodynamic comparison of two

calcium supplements in postmenopausal women. J Clin Phar-

macol. 2000;40(11):1237–44.

101. Harvey JA, Kenny P, Poindexter J, Pak CY. Superior calcium

absorption from calcium citrate than calcium carbonate using

external forearm counting. J Am Coll Nutr. 1990;9(6):583–7.

102. Pak CY, Avioli LV. Factors affecting absorbability of calcium

from calcium salts and food. Calcif Tissue Int. 1988;43(2):55–60.

103. Sakhaee K, Pak C. Superior calcium bioavailability of effer-

vescent potassium calcium citrate over tablet formulation of

calcium citrate after Roux-en-Y gastric bypass. Surg Obes Relat

Dis. 2011;. doi:10.1016/j.soard.2011.11.011.

104. Sakhaee K, Griffith C, Pak CY. Biochemical control of bone

loss and stone-forming propensity by potassium-calcium citrate

after bariatric surgery. Surg Obes Relat Dis. 2012;8(1):67–72.

doi:10.1016/j.soard.2011.05.001.

105. Rosen CJ, Brown S. Severe hypocalcemia after intravenous bis-

phosphonate therapy in occult vitamin D deficiency. N Engl J Med.

2003;348(15):1503–4. doi:10.1056/NEJM200304103481521.

Clinic Rev Bone Miner Metab

123