Skip to main content
Erschienen in: BMC Nephrology 1/2019

Open Access 01.12.2019 | Research article

Association of Constipation with risk of end-stage renal disease in patients with chronic kidney disease

verfasst von: Chung-Yen Lu, Yin-Cheng Chen, Yu-Wen Lu, Chih-Hsin Muo, Ray-E Chang

Erschienen in: BMC Nephrology | Ausgabe 1/2019

Abstract

Background

Chronic Kidney Disease (CKD) is a growing public health problem. Many risk factors were identified and interventions were applied accordingly, but the incidence of end-stage renal disease continued increasing. Some other risk factors may be ignored. Gut microbiota has been recognized as an important endogenous organ. The kidney-gut axis would contribute to gut dysbiosis, which might worsen CKD. Constipation, commonly seen in CKD, was one of the clinical presentation of gut dysbiosis. The clinical impact of constipation to CKD remains unknown. Our study aimed at assessing the risk of ESRD between CKD patients with and without constipation in a nationwide database.

Methods

We identified newly diagnosed cases of CKD without constipation history before in 2000–2011 from the Taiwan National Health Insurance database. Subjects who developed constipation later formed constipation group. The others without constipation matched by propensity score formed non-constipation group. The incidence rates and hazards of ESRD in patients with and without constipation by the end of 2013 were compared by using Cox proportional hazard models with a time-dependent variable.

Results

The incidences of ESRD per 1000 person-years were 22.9 for constipation group and 12.2 for non-constipation group, respectively. Cox proportional hazard models with a time-dependent variable revealed an adjusted hazard ratio of 1.90 (95% CI, 1.60–2.27). Compared to the CKD patients without constipation, adjusted hazard ratio for the CKD patients with laxatives < 33, 33–197 and ≥ 198 days per year were 0.45 (0.31–0.63), 1.85 (1.47–2.31) and 4.41 (3.61–5.39) respectively.

Conclusion

In a population of newly-diagnosed CKD patients, we observed that subjects with de novo constipation, as compared with non-constipation, have increased risk of developing ESRD. More severe constipation would increase the risk further.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12882-019-1481-0) contains supplementary material, which is available to authorized users.
Chung-Yen Lu and Yin-Cheng Chen contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

Chronic kidney disease (CKD) is a global health issue. If CKD couldn’t be adequately controlled, it would progress to end-stage renal disease (ESRD), which requires costly intervention of renal replacement therapy and will be a great burden to family and government [1]. There are many risk factors for CKD, including diabetes mellitus, hypertension, analgesics, herbs, kidney stones, and infection, etc. [28]. However, even under strict control, CKD may still progress. Some other unknown risk factors may be ignored.
In the near decades, human gut was found to harbor 1011 to 1012 microbiota which influence the nutrition, metabolism, and immune function of the host and has been recognized as an endogenous organ inside the body [912]. In healthy people, the gut microbiota interact harmoniously with the host in mutualistic relationship, so called symbiosis. However, many chronic diseases will disturb the gut microbiome and turn symbiosis to dysbiosis, which was related to some complications and the worsening of the chronic disease [13]. The interaction between kidney and gut, so called kidney-gut axis, was one of the concerned [14]. In the “kidney-to-gut” side, CKD is associated with fluid overload with intestinal wall edema, accumulation of uremic toxin, decreased consumption of dietary fibers, oral use of phosphate binders and/or iron, and so on. These factors all contribute to intestinal dysbiosis [12, 15]. Constipation is the most common gut abnormality in CKD patients. In the opposite “gut-to-kidney” side, gut microbiota also produce some uremia retention molecules, like indoxyl sulfate and p-cresyl sulfate, which are fully excreted by the normal kidney but would accumulate as kidney fails [16]. The increased levels of these metabolites are associated with chronic inflammation, increased cardiovascular mortality and the progression of CKD [1719]. The interaction between kidney and gut would be a vicious cycle that worsen the kidney further. Constipation might be an indicator of gut dysbiosis in CKD patients and suggested an ongoing vicious cycle. However, the constipation in CKD patients was usually ignored and not even thought as a risk factor of the progression of CKD.
Whether the CKD patients with constipation had poor profiles of gut microbiome inside was not known yet. We hypothesized CKD patients with constipation may suffer from rapid progression of kidney disease for the negative effects of poor profiles of gut microbiome. The current study tried to explore the relationship between CKD progression and constipation by analyzing a nationwide database.

Methods

Data source

We used the Longitudinal Health Insurance Database (LHID) 2000 [20], a medical claims database that includes one million subjects randomly selected from among people insured during 1996–2000 by the National Health Insurance (NHI) of Taiwan, to design the population-based retrospective cohort study. 96% of the total 23 million Taiwan’s residents were covered by the healthcare of NHI by the end of 1996 [21] and more than 99% by 2011 [22]. The LHID was for research purposes and represented the general population by age and sex distributions and included multiple claims files, like the expenditures and orders of ambulatory and inpatient care, the prescriptions of drugs, and registry for beneficiaries, which altogether provide information about the diagnoses and details of the utilization of healthcare. The diagnostic codes of the claims are recorded according to the International Classification of Diseases, 9th Revision, Clinical Modification (ICD-9-CM). According to Personal Information Protection Act, the identification of insurant was re-coded and this study was approved by the Institutional Review Board of the China Medical University Hospital (CMUH104-REC2–115(CR-1)) in Taiwan.

Study population

Within LHID 2000, those more than 20 years old and with new CKD diagnosis (CKD related ICD-9-CM codes shown in Additional file 1: Table S1) in at least three outpatient claims between 1 January 2000 and 31 December 2011 were collected as study population (n = 54052). The date of the third CKD diagnosis was the CKD date. The date of the diagnosis of constipation was the index date (detailed definition seen “Exposures to constipation”). 131 subjects participating health insurance less than 2 years before the CKD date were excluded. 23 subjects with ESRD diagnosis any time before the CKD date, and 1001 subjects with ESRD diagnosis any time before the index date, were excluded. Finally, 26780 patients with constipation history before their index date were excluded. A total of 26117 subjects were included in the data analysis (Fig. 1).

Exposures to constipation

We defined constipation group as patients who had at least two claims for constipation (ICD-9-CM 564.0x) or Laxatives prescription (ACT code A06AA, A06AB, A06AC, A06AD, A06AG, A06AH or A06AX) over 60 days apart within one year before their index date. The date of their second visit of a constipation outpatient clinic was the index date. In the cohort, those with constipation were 4035 and without constipation was 18797. The constipation group and non-constipation group, each consisting of 3909 subjects, were formed after matching by propensity score according to age, region, insurance salary, CKD-year, index year, concomitant diseases, and related medications (Fig. 1). Among constipation group, we further divided them into subgroups according to the duration of laxative use. For there were no insights or references about the relationships between the duration of constipation and ESRD, we thus divided the constipation group into 3 equal subgroups by the numbers of the subjects, which may represent mild, moderate, and severe constipation respectively, were done. The cut-off time was the lower third: < 33, the middle third: 33–197 and the upper third: ≥198 days per year. Besides, the different numbers of types (less than or equal to one type (≤ 1 type) and more than one type (> 1 type)) of laxatives used were compared too.

Patient characteristics

The demographic variables included age, sex, geographic region and urbanization level [23]. Two kinds of concomitant diseases were adopted. The first was the comorbidities related to the progression of CKD, including diabetes mellitus, hypertension, hyperlipidemia, acute coronary syndrome, cerebrovascular disease and chronic obstructive pulmonary disease. The second was the diseases predisposing constipation, including gastrointestinal tract malignancy, inflammatory bowel disease, hypothyroidism, Parkinson’s disease, and mytonic dystrophy. (Concomitant diseases ICD-9-CM codes shown in supplementary Table S1). Besides, these concomitant diseases were considered to be present if the diagnosis codes were recorded on at least one inpatient claim or two outpatient claims in the two years before the index date. Also two kinds of medications were adopted. The first was the medication which may cause nephrotoxicity, including non-steroid anti-inflammatory drugs (NSAIDs) and analgesics other than NSAIDs. The second was the medication which may cause constipation, including opioids, aluminum antacids, antidepressants, antihistamines, antispasmodics, anticonvulsants, antiarrythmics, anti-diarrheals, 5-HT3 receptor antagonists, beta-adrenoceptor antagonists, calcium channel blockers, diuretics, calcium and iron supplement (Related medications ATC classification system codes shown in Additional file 1: Table S2).

Outcome variable

The study end point was ESRD, which was defined by CKD plus chronic renal failure under regular dialysis (ICD-9-CM codes 585) from the registry for catastrophic illness patient database in Taiwan NHI. ESRD rate = ESRD events numbers / 1000 person-years. The ESRD rate in non-constipation group was the reference. The ESRD rate in the other groups was compared to the non-constipation.

Statistical analysis

The baseline characteristics of constipation group and non-constipation group were summarized using descriptive statistics. The follow-up duration started at the index date and ended at the occurrence of ESRD, withdrawal from NHI program, or the termination of this study on 31 December 2013, whichever came first. To account for the immortal time of constipation patients, we assessed the association between constipation and risk of developing ESRD using Cox proportional hazard models with a time-dependent variable for constipation group. For people in constipation group, their follow-up person-time were exactly classified into the non-constipation group, before the index date of the constipation, and the constipation group, after the index date. The Cox models yielded the hazard ratio (HR) and 95% confidence interval (CI) for ESRD in constipation group, with non-constipation group as the reference group. All statistical analyses were performed using SAS 9.4 software for windows (SAS Institute, Cary, NC, USA) and the significant level was set at 0.05 in two-tailed test.

Results

The demographics, socioeconomic status, concomitant diseases and related medications between the two groups with and without constipation were presented in Table 1. Among the 7818 newly diagnosed cases of CKD, 3909, or 50%, had constipation and the other 50% had no constipation during the follow-up time. Table 1 presented the patients’ characteristics in two years before index date. Data for demographics, socioeconomic status, concomitant diseases and related medications showed no difference in distribution between groups.
Table 1
Patients’ characteristics
Variable
Overall
Constipation
Non-constipation
p-value
Patient no.
N = 7818
N = 3909
N = 3909
 
Age, years (S.D.)
65.2
(14.3)
65.0
(15.0)
65.4
(13.5)
0.20
Female (%)
3540
(45.3)
1766
(45.2)
1774
(45.4)
0.86
Insurance amount, NTD (%)
0.77
Fixed premium or dependent
1519
(19.4)
765
(19.6)
754
(19.3)
 
< 20,000
3276
(41.9)
1623
(41.5)
1653
(42.3)
 
20,000–39,999
2391
(30.6)
1194
(30.5)
1197
(30.6)
 
≧39,999
632
(8.08)
327
(8.37)
305
(7.80)
 
Region (%)
0.87
 North
2779
((35.6)
1386
(35.5)
1393
(35.6)
 
 Center
1710
(21.9)
856
(21.9)
854
(21.9)
 
 South
2608
(33.4)
1296
(33.2)
1312
(33.6)
 
 East
721
(9.22)
371
(9.49)
350
(8.95)
 
Urbanization (%)
0.88
Urban
4403
(56.3)
2212
(56.6)
2191
(56.1)
 
 Satellite
2529
(32.4)
1259
(32.2)
1270
(32.5)
 
 Rural
886
(11.3)
438
(11.2)
448
(11.5)
 
Comorbidities (%)
 Acute coronary syndrome
371
(4.75)
188
(4.81)
183
(4.68)
0.79
Diabetes
3735
(47.8)
1862
(47.6)
1873
(47.9)
0.80
Hypertension
5277
(67.5)
2623
(67.1)
2654
(67.9)
0.45
Hyperlipidemia
2919
(37.3)
1452
(37.2)
1467
(37.5)
0.73
COPD
1046
(13.4)
516
(13.2)
530
(13.6)
0.64
Cerebrovascular disease
631
(8.07)
343
(8.77)
288
(7.37)
0.02
Gastrointestinal tract cancer
227
(2.90)
113
(2.89)
114
(2.92)
0.95
Inflammatory bowel disease
68
(0.87)
31
(0.79)
37
(0.95)
0.46
Hypothyroidism
101
(1.29)
53
(1.36)
48
(1.23)
0.62
Parkinson’s disease
184
(2.35)
100
(2.56)
84
(2.15)
0.23
Mytonic dystophy
0
(0.00)
0
(0.00)
0
(0.00)
 
Medications (%)
 NSAIDs
6271
(80.2)
3121
(79.8)
3150
(80.6)
0.41
 Analgesic drugs other than NSAIDs
6429
(82.2)
3210
(82.1)
3219
(82.4)
0.79
 Aluminum antacids
873
(11.2)
424
(10.9)
449
(11.5)
0.37
 Antiarrythmics
471
(6.02)
237
(6.06)
234
(5.99)
0.89
 Anticonvulsants
1052
(13.5)
537
(13.7)
515
(13.2)
0.47
 Antidepressants
1271
(16.3)
646
(16.5)
625
(16.0)
0.52
 Antidiarrheal
1155
(14.8)
578
(14.8)
577
(14.8)
0.97
 Antihistamines
5590
(71.5)
2775
(71.0)
2815
(72.0)
0.32
 Antispasmodics
2780
(35.6)
1373
(35.1)
1407
(36.0)
0.42
 Beta-blockers
3277
(41.9)
1627
(41.6)
1650
(42.2)
0.60
 Calcium channel blockers
4390
(56.2)
2197
(56.2)
2193
(56.1)
0.93
 Calcium supplement
986
(12.6)
493
(12.6)
493
(12.6)
> 0.99
 Diuretics
3408
(43.6)
1694
(43.3)
1714
(43.9)
0.65
 Iron supplement
568
(7.27)
279
(7.14)
289
(7.39)
0.66
 Opioids
1630
(20.9)
821
(21.0)
809
(20.7)
0.74
 Serotonin (5HT3) antagonists
95
(1.22)
43
(1.10)
52
(1.33)
0.35
Propensity score, (S.D.)
0.30
(0.19)
0.30
(0.19)
0.30
(0.19)
> 0.99
Chi-square test and t-test
Duration between the date of index and CKD was 4.70 ± 3.41 years for overall, 4.74 ± 3.36 years for Constipation, and 4.67 ± 3.46 years for non- Constipation (t-test p = 0.31)
In follow-up to the end of 2013, 371 patients (9.5%) experienced ESRD in those with constipation as compared to 182 (4.7%) in those without constipation, respectively (Table 2). Over the 13-year period, the incidences of ESRD per 1000 person-years were 22.9 for constipation group and 12.2 for non-constipation group. Figure 2 showed that there was statistically significant difference for cumulative incidences for ESRD between the patients with and without constipation (P < 0.0001). Cox proportional hazard models with a time-dependent variable revealed an adjusted HR of 1.90 (95% CI, 1.60–2.27) in the CKD patients with constipation (Table 2). Compared to the CKD patients without constipation, adjusted HR for the CKD patients with laxatives < 33, 33–197 and ≥ 198 days per year were 0.45 (0.31–0.63), 1.85 (1.47–2.31) and 4.41 (3.61–5.39) respectively. Adjusted HR for the CKD patients with laxatives ≤ 1 type and > 1 type during the study period were 2.08 (1.76–2.50) and 1.53 (1.19–1.95), respectively, compared to the non-constipation group.
Table 2
Incidence and hazard ratio for ESRD and associated risk factor
Variable
N
ESRD events
Person-years
*Rate
HR (95% CI)
p-value
Constipation
 No
3909
182
14906
12.21
Ref.
 
 Yes
3909
371
16232
22.86
1.90 (1.60–2.27)
< 0.0001
Constipation, duration
 No
3909
182
14906
12.21
Ref.
 
 With Laxatives, <  33 days per year
1291
36
7008
5.14
0.45 (0.31–0.63)
< 0.0001
 With Laxatives, 33–197 days per year
1291
123
5504
22.35
1.85 (1.47–2.31)
< 0.0001
 With Laxatives, ≥ 198 days per year
1327
212
3720
56.99
4.41 (3.61–5.39)
< 0.0001
Constipation, numbers of types of Laxatives
 No
3909
182
14906
12.21
Ref.
 
 With Laxatives, ≤ one type during the study period
2844
277
10882
25.45
2.08 (1.76–2.50)
< 0.0001
 With Laxatives, > one type during the study period
1065
94
5349
17.57
1.53 (1.19–1.95)
0.0008
*Rate: ESRD events per 1000 person-years

Discussions

To investigate the impact of kidney-gut axis, our study compared the difference of occurrence of ESRD between CKD patients with newly developed constipation and without constipation. The baseline of demographic and socioeconomic status was matched by propensity scores. The well-known comorbidities related to the progression of CKD and the nephrotoxic medications were also included and matched. Furthermore, the concomitant diseases and medications that might cause constipation were also added and matched. The 2 groups, CKD with constipation and CKD without constipation, were fairly matched and then followed longitudinally from 2000 to 2013. The ESRD events was 371, or 22.9 per 1000 person-year, in CKD with constipation and 182, or 4.7 per 1000 person-year, in CKD without constipation respectively. The cumulative incidence for ESRD in CKD with constipation was significantly higher than those without. The adjusted HR for CKD with constipation was 1.90 (95% CI, 1.60–2.27). The result showed de novo constipation in CKD patients increased the risk of the development of ESRD. To clarify the relationship between CKD and constipation, we further analyzed the incidence of ESRD in constipation patients with different duration of laxatives use per year and different numbers of types of laxatives. Compared to the CKD patients without constipation, adjusted HR for the CKD patients with laxatives < 33, 33–197 and ≥ 198 days per year were 0.45 (0.31–0.63), 1.85 (1.47–2.31) and 4.41 (3.61–5.39) respectively. For mild constipation, especially those who nearly did not need laxatives (laxatives < 33 days), the HR (0.45) of ESRD did not increase. However, for obvious constipation, with laxative use more than 33 days per year, the HR (1.85 for 33–197 and 4.41 for ≥198) of ESRD significant higher than non-constipation group. For those with even longer duration of laxatives use, more than 198 days per year, the HR (4.41) was higher than those with laxative use between 33 and 197 days per year (1.85). The result showed the mild constipation, with nearly no laxatives needed, would not increase the risk of ESRD; however, the obvious constipation, with laxatives use more than one month per year, would increase the risk of ESRD. Furthermore, more severe constipation, with laxatives use more than 6 months per year, would bring even higher risk of ESRD. There seemed to be a dose-effect relationship between CKD and constipation, which consolidated our hypothesis. The adjusted HR for the CKD patients with laxatives less than or equal to one type (≤ 1 type) and more than one type (> 1 type) during the study period were 2.08 (1.76–2.50) and 1.53 (1.19–1.95), respectively, compared to the non-constipation group. Both ≤ 1 type and > 1 type laxatives showed significantly increased HR, but the > 1 type did not have higher HR than ≤ 1 type. Combining the analysis above, the duration of laxative use should account more in the risk of ESRD than the different numbers of types of laxatives use.
The main limitation of this study is that we cannot rule out the possibility of residual confounding by unmeasured factors such as lifestyle and behavioral factors such as diet, smoking, alcohol drinking and exercise. Otherwise, we tried to reduce the confounders related to the progression of CKD and the development of constipation. To reduce the confounders related to the progression of CKD, several associated comorbidities, including acute coronary syndrome, diabetes, hypertension, hyperlipidemia, COPD and cerebrovascular disease, were added to the propensity score-matching models (Table 1). Previous use of NSAIDs and analgesic drugs other than NSAIDs were also added to matching. To reduce the confounders related to the development of constipation, the concomitant diseases and related medications which may cause constipation were also added into the matching. We tried to find if the de novo constipation per se increased the risk of ESRD in CKD patients.
Constipation status and severity was reported as a risk factor of incident CKD and ESRD [24]. It suggested healthy people needed to monitor their defecation status to maintain kidney health. Our study focused on the CKD people and suggested they should maintain good bowel habit to avoid de novo constipation which might precipitate the CKD progression. Moreover, even the constipation could be controlled by the laxatives, but the erroneous gut microbiome, that caused the negative effect to kidney via kidney-gut axis, might not be changed. Therefore, some other interventions, like the use of probiotics and prebioitics [2527], smart bacteriae [28], and high-fiber diet [29], or the application of toxin-absorbing agents [30, 31] should be considered to correct this error. It is also relevant to define the specific microbiome profiles associated with CKD and ESRD to give the precise therapy. Future studies should explore the profiles of microbiome in CKD and the interaction between them.

Conclusions

Our longitudinal nationwide study showed that de novo constipation in CKD patients was a risk factor in progression to ESRD. The obvious constipation, with laxatives needed, increased the risk. The more severe constipation, with the longer duration of laxatives needed, increased the risk further. The patency issue of bowel should not be ignored in CKD patients. Dietary modification and some other interventions should be tried to avoid constipation. Avoidance or even shortened intervals of constipation might be helpful to lessen the progression of CKD.

Acknowledgements

This study is based on data from the National Health Insurance Research Database provided by the Bureau of National Health Insurance, Department of Health and managed by National Health Research Institutes. The interpretation and conclusions contained herein do not represent those of Bureau of National Health Insurance, Department of Health or National Health Research Institutes.
This study was approved by the Institutional Review Board of the China Medical University Hospital (CMUH104-REC2–115(CR-1)) in Taiwan. Written consent from study objects was not required and approved by the institutional review board, as the LHID 2000 consists of de-identified secondary data for research purposes.
not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Thomas B, Wulf S, Bikbov B, Perico N, Cortinovis M, Courville de Vaccaro K, Flaxman A, Peterson H, Delossantos A, Haring D, Mehrotra R, Himmelfarb J, Remuzzi G, Murray C, Naghavi M. Maintenance Dialysis throughout the world in years 1990 and 2010. J Am Soc Nephrol. 2015;26:2621–33.CrossRef Thomas B, Wulf S, Bikbov B, Perico N, Cortinovis M, Courville de Vaccaro K, Flaxman A, Peterson H, Delossantos A, Haring D, Mehrotra R, Himmelfarb J, Remuzzi G, Murray C, Naghavi M. Maintenance Dialysis throughout the world in years 1990 and 2010. J Am Soc Nephrol. 2015;26:2621–33.CrossRef
2.
Zurück zum Zitat Orchard TJ, Dorman JS, Maser RE, Becker DJ, Drash AL, Ellis D, LaPorte RE, Kuller LH. Prevalence of complications in IDDM by sex and duration. Pittsburgh epidemiology of diabetes complications study II. Diabetes. 1990;39:1116–24.CrossRef Orchard TJ, Dorman JS, Maser RE, Becker DJ, Drash AL, Ellis D, LaPorte RE, Kuller LH. Prevalence of complications in IDDM by sex and duration. Pittsburgh epidemiology of diabetes complications study II. Diabetes. 1990;39:1116–24.CrossRef
3.
Zurück zum Zitat Tuttle KR, Bakris GL, Bilous RW, Chiang JL, de Boer IH, Goldstein-Fuchs J, Hirsch IB, Kalantar-Zadeh K, Narva AS, Navaneethan SD, Neumiller JJ, Patel UD, Ratner RE, Whaley-Connell AT, Molitch ME. Diabetic kidney disease: a report from an ADA consensus conference. Am J Kidney Dis. 2014;64:510–33.CrossRef Tuttle KR, Bakris GL, Bilous RW, Chiang JL, de Boer IH, Goldstein-Fuchs J, Hirsch IB, Kalantar-Zadeh K, Narva AS, Navaneethan SD, Neumiller JJ, Patel UD, Ratner RE, Whaley-Connell AT, Molitch ME. Diabetic kidney disease: a report from an ADA consensus conference. Am J Kidney Dis. 2014;64:510–33.CrossRef
4.
Zurück zum Zitat Zucchelli P, ZuccalàA. Primary hypertension--how does it cause renal failure? Nephrol Dial Transplant 1994;9:223–225. Zucchelli P, ZuccalàA. Primary hypertension--how does it cause renal failure? Nephrol Dial Transplant 1994;9:223–225.
5.
Zurück zum Zitat Jun M, Hemmelgarn BR. Strategies for BP control in developing countries and effects on kidney function. Clin J Am Soc Nephrol. 2016;11:932–4.CrossRef Jun M, Hemmelgarn BR. Strategies for BP control in developing countries and effects on kidney function. Clin J Am Soc Nephrol. 2016;11:932–4.CrossRef
6.
Zurück zum Zitat Buckalew VM Jr, Schey HM. Renal disease from habitual antipyretic analgesic consumption: an assessment of the epidemiologic evidence. Medicine (Baltimore). 1986;65:291–303.CrossRef Buckalew VM Jr, Schey HM. Renal disease from habitual antipyretic analgesic consumption: an assessment of the epidemiologic evidence. Medicine (Baltimore). 1986;65:291–303.CrossRef
7.
Zurück zum Zitat Levey AS, Atkins R, Coresh J, Cohen EP, Collins AJ, Eckardt KU, Nahas ME, Jaber BL, Jadoul M, Levin A, Powe NR, Rossert J, Wheeler DC, Lameire N, Eknoyan G. Chronic kidney disease as a global public health problem: approaches and initiatives - a position statement from kidney disease improving global outcomes. Kidney Int. 2007;72:247–59.CrossRef Levey AS, Atkins R, Coresh J, Cohen EP, Collins AJ, Eckardt KU, Nahas ME, Jaber BL, Jadoul M, Levin A, Powe NR, Rossert J, Wheeler DC, Lameire N, Eknoyan G. Chronic kidney disease as a global public health problem: approaches and initiatives - a position statement from kidney disease improving global outcomes. Kidney Int. 2007;72:247–59.CrossRef
8.
Zurück zum Zitat Luyckx VA. Nephrotoxicity of alternative medicine practice. Adv Chronic Kidney Dis. 2012;19:129–41.CrossRef Luyckx VA. Nephrotoxicity of alternative medicine practice. Adv Chronic Kidney Dis. 2012;19:129–41.CrossRef
9.
Zurück zum Zitat Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006;124:837–48.CrossRef Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006;124:837–48.CrossRef
10.
Zurück zum Zitat Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science. 2005;307:1915–20.CrossRef Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science. 2005;307:1915–20.CrossRef
11.
Zurück zum Zitat Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient environment of the mammalian intestine. Annu Rev Nutr. 2002;22:283–307.CrossRef Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient environment of the mammalian intestine. Annu Rev Nutr. 2002;22:283–307.CrossRef
12.
Zurück zum Zitat Ramezani A, Raj DS. The gut microbiome, kidney disease, and targeted interventions. J Am Soc Nephrol. 2014;25:657–70.CrossRef Ramezani A, Raj DS. The gut microbiome, kidney disease, and targeted interventions. J Am Soc Nephrol. 2014;25:657–70.CrossRef
13.
Zurück zum Zitat Carding S, Verbeke K, Vipond DT, Corfe BM, Owen LJ. Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis. 2015;26:261–91. Carding S, Verbeke K, Vipond DT, Corfe BM, Owen LJ. Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis. 2015;26:261–91.
14.
Zurück zum Zitat Pahl MV, Vaziri ND. The chronic kidney disease - colonic Axis. Semin Dial. 2015;28:459–63.CrossRef Pahl MV, Vaziri ND. The chronic kidney disease - colonic Axis. Semin Dial. 2015;28:459–63.CrossRef
15.
Zurück zum Zitat Chaves LD, McSkimming DI, Bryniarski MA, Honan AM, Abyad S, Thomas SA, Wells S, Buck M, Sun Y, Genco RJ, Quigg RJ, Yacoub R. Chronic kidney disease, uremic milieu, and its effects on gut bacterial microbiota dysbiosis. Am J Physiol Renal Physiol. 2018;315:F487–502.CrossRef Chaves LD, McSkimming DI, Bryniarski MA, Honan AM, Abyad S, Thomas SA, Wells S, Buck M, Sun Y, Genco RJ, Quigg RJ, Yacoub R. Chronic kidney disease, uremic milieu, and its effects on gut bacterial microbiota dysbiosis. Am J Physiol Renal Physiol. 2018;315:F487–502.CrossRef
16.
Zurück zum Zitat Vanholder R, De Smet R, GlorieuxG, Argilés A, Baurmeister U, Brunet P, Clark W, Cohen G, De Deyn PP, Deppisch R, Descamps-Latscha B, Henle T, Jörres A, Lemke HD, Massy ZA, Passlick-Deetjen J, Rodriguez M, Stegmayr B, Stenvinkel P, Tetta C, Wanner C, Zidek W. European uremic toxin work group (EUTox): review on uremic toxins: classification, concentration, and interindividual variability. Kidney Int 2003;63:1934–1943.CrossRef Vanholder R, De Smet R, GlorieuxG, Argilés A, Baurmeister U, Brunet P, Clark W, Cohen G, De Deyn PP, Deppisch R, Descamps-Latscha B, Henle T, Jörres A, Lemke HD, Massy ZA, Passlick-Deetjen J, Rodriguez M, Stegmayr B, Stenvinkel P, Tetta C, Wanner C, Zidek W. European uremic toxin work group (EUTox): review on uremic toxins: classification, concentration, and interindividual variability. Kidney Int 2003;63:1934–1943.CrossRef
17.
Zurück zum Zitat Barreto FC, Barreto DV, Liabeuf S, Meert N, Glorieux G, Temmar M, Choukroun G, Vanholder R, Massy ZA. European uremic toxin work group (EUTox): serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients. Clin J Am Soc Nephrol. 2009;4:1551–8.CrossRef Barreto FC, Barreto DV, Liabeuf S, Meert N, Glorieux G, Temmar M, Choukroun G, Vanholder R, Massy ZA. European uremic toxin work group (EUTox): serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients. Clin J Am Soc Nephrol. 2009;4:1551–8.CrossRef
18.
Zurück zum Zitat Meijers BK, Claes K, Bammens B, de Loor H, Viaene L, Verbeke K, Kuypers D, Vanrenterghem Y, Evenepoel P. P-cresol and cardiovascular risk in mild-to-moderate kidney disease. Clin J AmSoc Nephrol. 2010;5:1182–9.CrossRef Meijers BK, Claes K, Bammens B, de Loor H, Viaene L, Verbeke K, Kuypers D, Vanrenterghem Y, Evenepoel P. P-cresol and cardiovascular risk in mild-to-moderate kidney disease. Clin J AmSoc Nephrol. 2010;5:1182–9.CrossRef
19.
Zurück zum Zitat Wu IW, Hsu KH, Lee CC, Sun CY, Hsu HJ, Tsai CJ, Tzen CY, Wang YC, Lin CY, Wu MS. P-Cresyl sulphate and indoxyl sulphate predict progression of chronic kidney disease. Nephrol Dial Transplant. 2011;26:938–47.CrossRef Wu IW, Hsu KH, Lee CC, Sun CY, Hsu HJ, Tsai CJ, Tzen CY, Wang YC, Lin CY, Wu MS. P-Cresyl sulphate and indoxyl sulphate predict progression of chronic kidney disease. Nephrol Dial Transplant. 2011;26:938–47.CrossRef
21.
Zurück zum Zitat Lu JF, Hsiao WC. Does universal health insurance make health care unaffordable? Lessons from Taiwan. Health Aff (Millwood). 2003;22:77–88.CrossRef Lu JF, Hsiao WC. Does universal health insurance make health care unaffordable? Lessons from Taiwan. Health Aff (Millwood). 2003;22:77–88.CrossRef
23.
Zurück zum Zitat Liu CYHY, Chuang YL, et al. Incorporating development stratification of Taiwan townships into sampling design of large scale health interview survey. J Health Manag. 2006;4:1–22. Liu CYHY, Chuang YL, et al. Incorporating development stratification of Taiwan townships into sampling design of large scale health interview survey. J Health Manag. 2006;4:1–22.
24.
Zurück zum Zitat Sumida K, Molnar MZ, Potukuchi PK, Thomas F, Lu JL, Matsushita K, Yamagata K, Kalantar-Zadeh K, Kovesdy CP. J Am Soc Nephrol. 2017;28:1248–58.CrossRef Sumida K, Molnar MZ, Potukuchi PK, Thomas F, Lu JL, Matsushita K, Yamagata K, Kalantar-Zadeh K, Kovesdy CP. J Am Soc Nephrol. 2017;28:1248–58.CrossRef
25.
Zurück zum Zitat Ranganathan N, Patel B, Ranganathan P, Marczely J, Dheer R, Chordia T, Dunn SR, Friedman EA. Probiotic amelioration of azotemia in 5/6th nephrectomized Sprague-Dawley rats. Sci World J. 2005;5:652–60.CrossRef Ranganathan N, Patel B, Ranganathan P, Marczely J, Dheer R, Chordia T, Dunn SR, Friedman EA. Probiotic amelioration of azotemia in 5/6th nephrectomized Sprague-Dawley rats. Sci World J. 2005;5:652–60.CrossRef
26.
Zurück zum Zitat Chen L, Liu W, Li Y, Luo S, Liu Q, Zhong Y, Jian Z, Bao M. Lactobacillus acidophilus ATCC 4356 attenuates the atherosclerotic progression through modulation of oxidative stress and inflammatory process. Int Immunopharmacol. 2013;17:108–15.CrossRef Chen L, Liu W, Li Y, Luo S, Liu Q, Zhong Y, Jian Z, Bao M. Lactobacillus acidophilus ATCC 4356 attenuates the atherosclerotic progression through modulation of oxidative stress and inflammatory process. Int Immunopharmacol. 2013;17:108–15.CrossRef
27.
Zurück zum Zitat Meijers BK, De Preter V, Verbeke K, Vanrenterghem Y, Evenepoel P. P-Cresyl sulfate serum concentrations in haemodialysis patients are reduced by the prebiotic oligofructose-enriched inulin. Nephrol Dial Transplant. 2010;25:219–24.CrossRef Meijers BK, De Preter V, Verbeke K, Vanrenterghem Y, Evenepoel P. P-Cresyl sulfate serum concentrations in haemodialysis patients are reduced by the prebiotic oligofructose-enriched inulin. Nephrol Dial Transplant. 2010;25:219–24.CrossRef
28.
Zurück zum Zitat Pinero-Lambea C, Ruano-Gallego D, Fernandez LA. Engineered bacteria as therapeutic agents. Curr Opin Biotechnol. 2015;35:94–102.CrossRef Pinero-Lambea C, Ruano-Gallego D, Fernandez LA. Engineered bacteria as therapeutic agents. Curr Opin Biotechnol. 2015;35:94–102.CrossRef
29.
Zurück zum Zitat Krishnamurthy VM, Wei G, Baird BC, Murtaugh M, Chonchol MB, Raphael KL, Greene T, Beddhu S. High dietary fiber intake is associated with decreased inflammation and all-cause mortality in patients with chronic kidney disease. Kidney Int. 2012;81:300–6.CrossRef Krishnamurthy VM, Wei G, Baird BC, Murtaugh M, Chonchol MB, Raphael KL, Greene T, Beddhu S. High dietary fiber intake is associated with decreased inflammation and all-cause mortality in patients with chronic kidney disease. Kidney Int. 2012;81:300–6.CrossRef
30.
Zurück zum Zitat Schulman G, Berl T, Beck GJ, Remuzzi G, Ritz E, Arita K, Kato A, Shimizu M. Randomized placebo-controlled EPPIC trials of AST-120 in CKD. J Am Soc Nephrol. 2015;26:1732–46.CrossRef Schulman G, Berl T, Beck GJ, Remuzzi G, Ritz E, Arita K, Kato A, Shimizu M. Randomized placebo-controlled EPPIC trials of AST-120 in CKD. J Am Soc Nephrol. 2015;26:1732–46.CrossRef
31.
Zurück zum Zitat Schulman G, Berl T, Beck GJ, Remuzzi G, Ritz E, Shimizu M, Shobu Y, Kikuchi M. The effects of AST-120 on chronic kidney disease progression in the United States of America: a post hoc subgroup analysis of randomized controlled trials. BMC Nephrol. 2016;17:141.CrossRef Schulman G, Berl T, Beck GJ, Remuzzi G, Ritz E, Shimizu M, Shobu Y, Kikuchi M. The effects of AST-120 on chronic kidney disease progression in the United States of America: a post hoc subgroup analysis of randomized controlled trials. BMC Nephrol. 2016;17:141.CrossRef
Metadaten
Titel
Association of Constipation with risk of end-stage renal disease in patients with chronic kidney disease
verfasst von
Chung-Yen Lu
Yin-Cheng Chen
Yu-Wen Lu
Chih-Hsin Muo
Ray-E Chang
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Nephrology / Ausgabe 1/2019
Elektronische ISSN: 1471-2369
DOI
https://doi.org/10.1186/s12882-019-1481-0

Weitere Artikel der Ausgabe 1/2019

BMC Nephrology 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.