Skip to main content
Erschienen in: Gut Pathogens 1/2016

Open Access 01.12.2016 | Research

Case–control study of diarrheal disease etiology in individuals over 5 years in southwest China

verfasst von: Shun-Xian Zhang, Chun-Li Yang, Wen-Peng Gu, Lin Ai, Emmanuel Serrano, Pin Yang, Xia Zhou, Shi-Zhu Li, Shan Lv, Zhi-Sheng Dang, Jun-Hu Chen, Wei Hu, Li-Guang Tian, Jia-Xu Chen, Xiao-Nong Zhou

Erschienen in: Gut Pathogens | Ausgabe 1/2016

Abstract

Background

Acute diarrhea is one of the major public health problems worldwide. Most of studies on acute diarrhea have been made on infants aged below 5 years and few efforts have been made to identify the etiological agents of acute diarrhea in people over five, especially in China.

Methods

271 diarrhea cases and 149 healthy controls over 5 years were recruited from four participating hospitals between June 2014 and July 2015. Each stool specimen was collected to detect a series of enteric pathogens, involving five viruses (Rotavirus group A, RVA; Norovirus, NoV; Sapovirus, SaV; Astrovirus, As; and Adenovirus, Ad), seven bacteria (diarrheagenic Escherichia coli, DEC; non-typhoidal Salmonella, NTS; Shigella spp.; Vibrio cholera; Vibrio parahaemolyticus; Aeromonas spp.; and Plesiomonas spp.) and three protozoa (Cryptosporidium spp., Giardia lamblia, G. lamblia, and Blastocystis hominis, B. hominis). Standard microbiological and molecular methods were applied to detect these pathogens. Data was analyzed using Chi square, Fisher-exact tests and logistic regressions.

Results

The prevalence of at least one enteric pathogen was detected in 29.2% (79/271) acute diarrhea cases and in 12.1% (18/149) in healthy controls (p < 0.0001). Enteric viral infections (14.4%) were the most common in patients suffering from acute diarrhea, followed by bacteria (13.7%) and intestinal protozoa (4.8%). DEC (12.5%) was the most common causative agent in diarrhea cases, followed by NoV GII (10.0%), RVA (7.4%) and B. hominis (4.8%). The prevalence of co-infection was statistically higher (p = 0.0059) in the case group (7.7%) than in the healthy control (1.3%). RVA–NoV GII (3.0%) was the most common co-infection in symptomatic cases.

Conclusions

DEC was the most predominant pathogen in diarrhea cases, but it was largely overlooked because the lack of laboratory capacities. Because of the high prevalence of co-infections, it is recommended the urgent development of alternative laboratory methods to assess polymicrobial infections. Such methodological improvements will result in a better prevention and treatment strategies to control diarrhea illness in China.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13099-016-0141-1) contains supplementary material, which is available to authorized users.
Shun-Xian Zhang, Chun-Li Yang and Wen-Peng Gu contributed equally to this manuscript
Jia-Xu Chen and Xiao-Nong Zhou contributed equally to this manuscript
Abkürzungen
Ad
adenovirus
As
astrovirus
AWP
alkaline peptone water
B. hominis
Blastocystis hominis
CI
confidence interval
CFU
colony-forming unit-megakaryocyte
DEC
diarrheagenic E. coli
EAEC
enteroaggregative E. coli
EHEC
enterohemorrhagic E. coli
EIEC
enteroinvasive E. coli
EPEC
enteropathogenic E. coli
ETEC
enterotoxigenic E. Coli
G. lamblia
Giardia lamblia
MAC
macConkey agar
NoV
norovirus
NTS
non-typhoidal salmonella
OR
odd ratios
qPCR
quantitative PCR
RT-PCR
reverse transcription polymerase chain reaction
RVA
rotavirus A group
SaV
sapovirus
SD
standard deviation
SS
Salmonella–Shigella
TCBS
thiosulfate-citrate-bile salts-sucrose
WHO
World Health Organization
XLD
xylose, lysine and deoxycholate agar

Background

Diarrheal illness is still a serious public health problem that particularly affects individuals in middle and low income countries [1]. Diarrhea is still a major reason of attendance at health services and one of the general causes for hospital admission [2]. In addition, 1,400,000 million deaths are caused by diarrhea across all age groups, of which 700,000 deaths are over 5 years [1, 2].
The main enteric pathogens include a wide range of bacteria (e.g. diarrheagenic Escherichia coli, DEC; non-typhoidal Salmonella, NTS; Shigella spp.; Vibrio cholera; Vibrio parahaemolyticus; Aeromonas spp.; Plesiomonas spp.; Campylobacter spp.), virus (e.g. rotavirus group A, RVA; norovirus, NoV; Sapovirus, SaV; astrovirus, As; adenovirus, Ad; enterovirus.) and enteric parasites (e.g. Cryptosporidium spp.; Giardia lamblia, G. lamblia; Entamoeba histolytica and Blastocystis hominis, B. hominis) [38].
Most researches of enteric pathogens on individuals with and without diarrhea have been largely based on a single or few pathogen species [911]. However, co-infection is a common prevalence in diarrhea cases in such communities with poor food hygiene, low sanitation and contaminated water (35.0, 20.1, 13.0%, respectively) [6, 12, 13]. Co-infection, however, are also common in healthy patients (8.0, 5.3, 0.8%, respectively) [6, 12, 13]. Co-infection is of particular human health importance because pathogen species can interact within the host. Interactions within the host can have either positive or negative effects on each of the co-infecting enteric pathogen species. Under positive enteric pathogen interactions, diarrheal disease transmission and progression are enhanced [6, 12, 14, 15].
Infectious diarrhea is still one of the important public health problems in China. The reported infectious diarrhea is up to 70,000,000, and the reported incidence of infectious diarrhea is 55.9/10,000,000 annually in China listed by China Information System for Diseases Control and Prevention. Diarrheal illness incidence is located in top three of 39 notifiable infectious diseases [11, 16]. However, in many medical institutions, the lack of clinical microbiology laboratories and detection capabilities hamper the detection of etiological agents of gastroenteritis. As result, etiology of gastroenteritis in China is achieved in less than 5.0% of patients [11]. In addition, most of the diarrhea studies have been limited to children under 5 years and either bacterial or viral species [11, 17, 18]. Hence, the aim of the study was twofold: one was to investigate the etiology of diarrhea cases in people over 5 years and to assess patterns of co-infection among virus, bacteria and protozoa in patients from southwest China. This study will contribute to the effective control of acute diarrhea in the country.

Methods

Subjects of this study

Acute gastroenteritis patients were defined as those who had diarrhea over three times within 24 h with abnormal stool specimens (e.g. mucus stool, watery stool, loose stool or bloody stool) in accordance with the WHO standard [19]. Non-diarrheal subjects were defined as those who had no history of diarrhea symptom before 14 days and were recruited at the same time as diarrheal subjects.

Specimen and data collection

The stool specimens were collected from acute diarrhea cases and healthy controls over 5 years in outpatient from four sentinel hospitals as follows: The First people’s Hospital of Yunnan Province, Kunming Children’s Hospital, The Pushan Community Health Service Center in Kunming, The First People’s Hospital of Yunnan Province, and The First Affiliated Hospital of Kunming Medical University. A sterile sampling cup was applied to collect stool sample, with the criterion that each stool must be greater than 3 g or 3 mL, then each stool specimen was delivered to the laboratory of Yunnan Provincial Center for Disease Control and Prevention in Cary-Blair (C-B) culture medium (Oxoid Ltd, Basingstoke, UK) within 12 h. The clinical (e.g. fever, abdominal pain, nausea, vomiting, dehydration and tenesmus) and basic epidemiological data (e.g. sex, age, residence and season) was collected with structured questionnaire by doctors or nurses. The present study was conducted from July 2014 to June 2015.

Laboratory test for enteric pathogens

Each stool sample was divided into three aliquots (Additional file 1). The first one was used for isolating, culturing and identifying bacterial (DEC, NTS, Shigella spp., Vibrio cholera, Vibrio parahaemolyticus, Aeromonas spp. and Plesiomonas spp.), the second one detect viral pathogens (RVA; NoV; astrovirus As, and Adenovirus, Ad), and last to assess intestinal protozoa infection (Cryptosporidium spp., G. lamblia and B. hominis).

Bacterial detection

MacConkey agar (MAC, Oxoid Ltd, Basingstoke, UK) was used for culturing DEC, which was divided into five subtypes by their virulence genetic as following: enteroaggregative E. coli (EAEC), enterotoxigenic E. coli (ETEC), enteropathogenic E. coli (EPEC), enteroinvasive E. coli (EIEC) and enterohaemorrhagic E. coli (EHEC). The DEC subtypes were examined with quantitative PCR based on the previous literatures (Table 1) [20, 21]. Each stool sample was inoculated into the selenite brilliant green sulfa enrichment broth (Oxoid Ltd, Basingstoke, UK) for enrichment and then inoculated it onto Salmonella–Shigella agar (Oxoid Ltd, Basingstoke, UK) to detect NTS. In addition, each stool specimen was inoculated directly onto Salmonella–Shigella agar (Oxoid Ltd, Basingstoke, UK) to find Shigella spp. Moreover, each sample was inoculated onto alkaline peptone water (Oxoid Ltd, Basingstoke, UK) for enrichment, and then inoculated onto thiosulfate-citrate-bile salts-sucrose agar (Oxoid Ltd, Basingstoke, UK) to detect Vibrio cholera, Vibrio parahaemolyticus, Aeromonas spp. and Plesiomonas spp. For suspicious NTS, Shigella spp., Vibrio cholera, Vibrio parahaemolyticus, Aeromonas spp., and Plesiomonas spp. colonies. The systematic biochemical identification of was performed using the VITEK® 2 Compact instrument (bioMerieux, Marcyl’Etoile, France). Detailed detection procedures are found in references [11, 17].
Table 1
The primers and reactions condition applied to detect enteric pathogens in this study
Enteric pathogens
Target gene
Primer (5′–3′)
Amplicon sizes (bp)
Remarks
Source
EPEC
eae
CCACGGTTTATCAAACTGATAACG
105
Each stool specimen was inoculated to MAC media to culture DEC at 37 °C for 18 h, And then ten putative DEC colonies were selected to mix with 150 μL water to extract DNA at 100 °C for 10 min, and then the 20 μL volume of qPCR system is composed of 10 μL master mix (Takara Bio Inc, Shiga, Japan), 1 μL forward primer (10 μmol), 1 μL reverse primer (10 μmol), 1 μL DNA template and 7 μL H2O. The cycling conditions for each subtype DEC was 95 °C for 5 min, 40 cycles of 95 °C for 5 s, 60 °C for 30 s. The fluorescence recorded was at the annealing stage
[20, 21]
EHEC
stx1
ACTTCTCGACTGCAAAGACGTATG
132
ACAAATTATCCCCTGAGCCACTATC
stx2
CCACATCGGTGTCTGTTATTAACC
93
GGTCAAAACGCGCCTGATAG
ETEC
elt
TTCCCACCGGATCACCAA
62
CAACCTTGTGGTGCATGATGA
estA
CCTTTCGCTCAGGATGCTAAAC
128
CAGTAATTGCTACTATTCATGCTTTCAG
estB
CTTTCCCCTCTTTTAGTCAGTCAACT
137
GCAGTAAAATGTGTTGTTCATATTTTCTG
EAEC
aggR
CAGCGATACATTAAGACGCCTAAAG
116
CGTCAGCATCAGCTACAATTATTCC
EIEC
ipaH
ACCATGCTCGCAGAGAAACT
175
TCAGTACAGCATGCCATGGT
RVA
VP6
GACGGVGCRACTACATGGT
382
RVA, NoV GI, NoV GII, SaV and As were RNA viruses, complementary DNA (cDNA) was synthesized using a random primer (Takara Bio Inc, Shiga, Japan) at 55 °C for 1.5 h, followed by 100 °C for 10 min, and holding at 4 °C. The reaction condition of RVA was 94 °C for 5 min, followed by 40 cycles at 94 °C for 1 min, 42 °C for 1 min, 72 °C for 1 min, and with final extension at 72 °C for 10 min. Multiplex RT-PCR was used to detect the presence of NoV GI, NoV GII, and SaV, the thermal profile consisted of 94 °C for 5 min, 40 cycles of 94 °C for 70 s, 49 °C for 70 s, and 72 °C for 1 min, followed by 72 °C for 10 min. The thermal profile of As was 94 °C for 5 min, 40 cycles of 94 °C for 30 s, 55 °C for 30 s, and 72 °C for 1 min, followed by 72 °C for 10 min
[22]
GTCCAATTCATNCCTGGTGG
NoV GI
NoV GII
SaV
Polymerase
TGACGATTTCATCATCACCATA
331/319
[23]
TGACGATTTCATCATCCCCGTA
GATTACTCCAGGTGGGACTCCAC
GATTACTCCAGGTGGGACTCAAC
GATTACTCCAGGTGGGATTCAAC
GATTACTCCAGGTGGGATTCCAC
As
Capsid
CAACTCAGGAAACAGGGTGT
449
[24]
TCAGATGCATTGTCATTGGT
Ad
Hexon
TTCCCCATGGCICAYAACAC
482
The thermal profile was 94 °C for 5 min, 40 cycles of 94 °C for 30 s, 55 °C for 30 s, and 72 °C for 1 min, followed by 72 °C for 10 min
[25]
CCCTGGTAKCCRATRTTGTA
Blastocistis hominis
SSU-rRNA
CGAATGGCTCATTATATCAGTT
260
The thermal profile was 94 °C for 5 min, 40 cycles of 94 °C for 30 s, 53 °C for 30 s, and 72 °C for 1 min, followed by 72 °C for 10 min
[26]
TCTTCGTTACCCGTTACTGC
Cryptosporidium spp.
18S-rRNA
TTCTAGAGCTAATACATGCG
 
The primary cycle consisted of 94 °C for 5 min, 35 cycles of 94 °C for 50 s, 55 °C for 1 min and 72 °C for 90 s, followed by 72 °C for 10 min, the annealing step for a second reaction was 58 °C
[27]
CCCATTTCCTTCGAAACAGGA
 
GGAAGGGTTGTATTTATTAGATAAAG
840
CTCATAAGGTGCTGAAGGAGTA
Giardia lamblia
Tim
AAATIATGCCTGCTCGTCG
 
The thermal profile of first round was 94 °C for 1 min, 53 °C for 1 min, and 72 °C for 1 min, followed by 72 °C for 10 min. A second reaction was carried out similarly
[28]
CAAACCTTITCCGCAAACC
 
CCCTTCATCGGIGGTAACTT
530
GTGGCCACCACICCCGTGCC
DEC is composed of EAEC, EPEC, EIEC, ETEC and EHEC in this study, the judging standard of subtypes of DEC according to qPCR was: EPEC: eae+; EAEC: aggR+; EIEC: ipaH+; EHEC: eae+, and (stx1+; and/or stx2+); ETEC: hlt+, and/or estA, and/or estB+

Virus detection

Nucleic Acid was extracted from each stool specimen (15% wt/vol or vol/vol suspension) with QIAamp Viral RNA Kit (Qiagen, Hilden, Germany). The reverse transcription-polymerase chain reaction (RT-PCR) was applied to detected RVA [22], NoV (GI, GII) [23] and As [24]. For RT, the viral RNA was reverse transcribed with PrimeScript™ RT reagent Kit (Takara Bio Inc, Shiga, Japan). Ad was found using PCR [25] (Table 1).

Enteric protozoan detection

The genomic DNA of Cryptosporidium spp., G. lamblia and B. hominis was extracted from each stool sample with QIAamp DNA stool mini kit (Qiagen, Hilden, Germany) according to the manufacturers’ protocol. The conventional PCR was applied to detect B. hominis [26], the nested PCR was used to detect Cryptosporidium spp. [27] and G. lamblia [28] (Table 1).

Data analysis

Data analysis was performed by IBM SPSS software (version 19.0 for Windows, Armonk, NY). Odds ratio (OR) and 95% CIs of categorical variables were calculated using two tailed Chi square or Fisher’s exact tests. Quantitative variable was described as mean, median, standard deviation or interquartile range (IQR), among which the median or mean of quantitative variable was compared by rank-sum test, analysis of variance or t test. Logistic regression was performed to find the relationship between diarrhea illness and various enteric pathogens. Single etiology was selected according to bivariate analysis with p < 0.20. Significant difference was taken as the level of p < 0.05 with two-tailed test.

Results

Basic information and clinical symptoms

From July 2014 to June 2015, 420 subjects were recruited for this study, which including 271 diarrhea cases and 149 healthy controls over 5 years. The male-to-female ratio was 0.964 in diarrhea cases and 0.961 in healthy controls (χ2 < 0.001, p = 0.987), respectively. The median age was 40.0 years in acute diarrhea cases and 41.4 years in non-diarrheal group(t = 0.817, p = 0.414). The diarrhea cases from urban areas accounted for 67.9%, and the non-diarrhea patients accounted for 66.4% (χ2 = 1.240, p = 0.538). The subjects in the 5–15 years age group was 64.5% in diarrhea cases and 63.1% in healthy controls (χ2 = 0.767, p = 0.681). The most frequent clinical symptom was nausea (n = 91, 33.6%) in diarrhea cases, and other common symptoms included abdominal pain (n = 73, 26.9%), vomiting (n = 58, 21.4%) and fever (n = 22, 8.1%). Mucus stool (n = 173, 63.8%) was the most common stool type in diarrhea cases, followed by watery stool (n = 70, 25.8%) and other types of stool (n = 28, 10.3%) (Table 2). The frequency of diarrhea was 5.8 times in acute diarrhea cases within 24 h (Additional file 2).
Table 2
Basic information and clinical characteristics of 271 acute diarrhea cases and 149 controls over 5 years
Characteristic
Diarrhea
Control
n (%)
n (%)
n
271
149
Age
 5–15 years
21 (7.7)
9 (6.0)
 15–50 years
175 (64.6)
94 (63.1)
 ≥50 years
75 (27.7)
46 (30.9)
Sex
 Male
133 (49.1)
73 (49.0)
 Female
138 (50.9)
76 (51.0)
Residence
 Urban
184 (67.9)
99 (66.4)
 Rural–urban fringe zone
68 (25.1)
35 (23.5)
 Rural
19 (7.0)
15 (10.1)
Seasons
 Spring (Feb–Apr)
87 (32.1)
32 (21.5)
 Summer (May–Jul)
65 (24.0)
42 (28.2)
 Autumn (Aug–Oct)
59 (25.5)
45 (30.2)
 Winter (Nov–Jan)
50 (18.5)
30 (37.5)
Symptom
 Fever (>37.3 °C)
23 (8.5)
 Abdominal pain
73 (26.9)
 Nausea
91 (33.6)
 Vomiting
58 (21.4)
 Dehydration
3 (1.1)
 Tenesmus
5 (1.8)
Diarrhea
 
 Watery stool
70 (25.8)
 Mucus stool
173 (63.8)
 Other stool
28 (10.3)
SD represent for standard deviation. Kunming city (25º 02′ 20″ N, 102º 43′05″ E, 1891 m.a.s.l.) has a humid subtropical climate of moderate seasonality characterized by a mild (mean temperature = 11.4 °C, min = 8, max = 15) and dry (mean precipitation = 33.4 mm, min = 12, max = 89) autumn (Aug–Oct) and winter (Nov–Jan). Spring (Feb–Apr) and summer (May–Jul) are also mild (mean temperature = 23 °C, min = 19, max = 29) but wet (mean precipitation = 159.6 mm, min = 92, max = 206) seasons. The “–” symbol indicates the information can not be collected

The prevalence of enteropathogen in subjects with diarrhea or not

At least one enteropathogen was isolated from 79 (29.2%) of 271 acute diarrhea cases and 18 (12.1%) of 149 healthy controls (χ2 = 15.774, p < 0.0001). The overall prevalence of bacterial pathogen and viral pathogen in diarrhea cases were higher than in healthy controls (χ2 = 11.327, p = 0.001; χ2 = 10.795, p = 0.001 respectively. Table 3). At least one intestinal protozoa was found in 4.8% (n = 13) of cases and 6.0% (n = 9) of controls (χ2 = 0.299, p = 0.584) (Table 3). In univariate analysis, Details of the enteric pathogens isolates are presented in Table 3, and according to that EAEC, NoV and RVA were more prevalent (χ2 = 7.061, p = 0.008; χ2 = 9.160, p = 0.002; χ2 = 7.061, p = 0.008 respectively) in diarrhea patients (7.4, 10.0, 7.4%, respectively) than in healthy controls (1.3, 2.0, 1.3%, respectively, Table 3). No statistical difference was observed between acute diarrhea patients and healthy subjects for EPEC, ETEC, NTS, Plesiomonas spp., SaV, As, B. hominis and Cryptosporidium spp. In addition, other enteric parasites were not detected in subjects with and without diarrhea (Table 3). However, the multivariate analysis showed that only RVA was an enteric pathogen associated with diarrhea. But EAEC and NoV GII did not relate with diarrheal illness among individuals over 5 years (Table 3).
Table 3
Enteric pathogens in the stool samples with diarrhea cases (n = 271) and healthy controls (n = 149) in Kunming, China
Enteropathogen
Diarrhea cases
n = 271
n (%)
Healthy controls
n = 149
n (%)
Univariate analysis
Multivariate analyses
p value
OR (95% CI)
p value
OR (95% CI)
At least one enteropathogen
79 (29.2)
18 (12.1)
p < 0.0001
3.00 (1.71–5.23)
At least one enteric bacterial pathogens
37 (13.7)
5 (3.4)
p = 0.001
4.55 (1.75–11.85)
 DEC
34 (12.5)
5 (3.4)
p = 0.002
4.13 (1.58–10.80)
  EAEC
20 (7.4)
2 (1.3)
p = 0.008
5.86 (1.35–25.41)
p = 0.198
5.95 (1.33–26.63)
  EPEC
15 (5.5)
3 (2.0)
p = 0.088
2.85 (0.81–10.01)
p = 0.107
2.86 (0.80–10.27)
  ETEC
1 (0.4)
0 (0.0)
  EIEC
0 (0.0)
0 (0.0)
  EHEC
0 (0.0)
0 (0.0)
 NTS
2 (0.7)
0 (0.0)
p = 0.541
 Plesiomonas spp.
1 (0.4)
0 (0.0)
 Vibrio parahaemolyticus
0 (0.0)
0 (0.0)
 Vibrio cholera
0 (0.0)
0 (0.0)
 Aeromonas spp.
0 (0.0)
0 (0.0)
 Shigella spp.
0 (0.0)
0 (0.0)
At least one enteric virus pathogens
39 (14.4)
6 (4.0)
p = 0.001
4.00 (1.66–9.70)
 NoV GII
27 (10.0)
3 (2.0)
p = 0.002
5.38 (1.60–18.06)
p = 0.0794
3.86 (0.85–17.48)
 RVA
20 (7.4)
2 (1.3)
p = 0.008
5.86 (1.35–25.41)
p = 0.0166
4.50 (1.31–15.43)
 NoV GI
1 (0.4)
0 (0.0)
 SaV
1 (0.4)
0 (0.0)
 As
0 (0.0)
1 (0.7)
p = 0.355
 Ad
0 (0.0)
0 (0.0)
  
At least one enteric parasite pathogens
13 (4.8)
9 (6.0)
p = 0.584
0.78 (0.33–1.88)
  
 B. hominis
13 (4.8)
9 (6.0)
p = 0.584
0.78 (0.33–1.88)
p = 0.412
0.68 (0.27–1.71)
 Cryptosporidium spp.
1 (0.4)
0 (0.0)
 Giardia lamblia
0 (0.0)
0 (0.0)
Including the co-infection of enteric pathogens in diarrhea cases and healthy subjects. The “–” symbol indicates the data can not be calculated
In diarrhea cases, DEC (12.5%, n = 34) was the most common pathogen, followed by NoV GII (10.0%, n = 27), RVA (7.0%, n = 20) and B. hominis (4.8%, n = 13).

Temporal distribution of enteric pathogen in diarrhea cases

The prevalence of EAEC, EPEC, RVA and B. hominis showed strong seasonal variations (Table 4). The detection rate of EAEC in summer was higher than in winter (p = 0.0045), and the prevalence of EPEC in summer was higher than in winter (p = 0.0156). RVA was mainly prevalent in autumn and winter (p = 0.0015), and the prevalence peak of B. hominis was summer (p < 0.0001). NoV GII was not statistically different in four seasons (χ2 = 3.359, p = 0.341).
Table 4
The seasonal characteristics of mainly enteric pathogen isolated from diarrhea cases
Enteropathogen
Spring
(Feb–Apr)
n = 87
n (%)
Summer
(May–Jul)
n = 65
n (%)
Autumn
(Aug–Oct)
n = 69
n (%)
Winter
(Nov–Jan)
n = 50
n (%)
χ2
p value
EAEC
1 (1.2)
9 (13.8)
8 (11.6)
2 (4.0)
p = 0.0045
EPEC
7 (8.0)
7 (10.8)
1 (1.4)
0 (0.0)
p = 0.0156
RVA
4 (4.6)
0 (0.0)
8 (11.6)
8 (16.0)
p = 0.0015
NoV
5 (5.7)
7 (10.8)
10 (14.5)
5 (10.0)
3.359
p = 0.341
B. hominis
0 (0.0)
7 (10.8)
6 (8.7)
0 (0.0)
p < 0.0001
Including the co-infection of any enteric pathogens in diarrhea cases. The “–” symbol indicates that data be calculated with Fisher-exact tests

Prevalence of enteric pathogens in diarrhea cases in different age group

Acute diarrhea cases were divided into different age groups, in which 21 (7.7%), 175 (64.6%) and 75 (27.7%) belong to age groups of 5–15, 15–50 and ≥50 years (Table 5). EPEC infection was the highest in the age group of 5–15 years (p = 0.031) (Table 5), but the prevalence of EAEC, RVA, NoV GII and B. hominis were not statistical difference among these three age groups (Table 5), respectively.
Table 5
Prevalence of enteric pathogens in diarrhea cases in different age groups
Enteropathogen
Total
n = 271
n (%)
5–15 years
n = 21
n (%)
15–50 years
n = 175
n (%)
≥50 years
n = 75
n (%)
χ2
p value
At least one enteropathogens
79 (29.2)
9 (42.9)
52 (29.7)
18 (24.0)
2.90
p = 0.234
At least one bacterium
37 (13.7)
5 (23.8)
27 (15.4)
5 (6.7)
5.41
p = 0.0668
At least one virus
39 (14.4)
4 (19.0)
23 (13.1)
12 (16.0)
0.748
p = 0.688
At least one parasite
13 (4.8)
1 (4.8)
10 (5.7)
2 (2.7)
p = 0.654
EAEC
20 (7.4)
1 (4.8)
16 (9.1)
3 (4.0)
2.56
p = 0.323
EPEC
15 (5.5)
4 (19.0)
9 (5.0)
2 (2.7)
p = 0.031
NoV
27 (10.0)
4 (19.0)
15 (8.6)
8 (10.7)
2.35
p = 0.309
RVA
20 (7.4)
2 (9.5)
13 (7.4)
5 (6.7)
0.198
p = 0.906
B. hominis
13 (4.8)
1 (4.8)
10 (5.7)
2 (2.7)
p = 0.654
Including the co-infection of any enteric pathogens in diarrhea cases. The “–” symbol indicates that data be calculated with Fisher-exact tests

Co-infection of enteric pathogen in diarrhea cases and healthy cases

In this study, the prevalence of co-infection with more than one enteric pathogens was higher than in healthy controls (Table 6, p = 0.0059, OR = 6.17, 95% CI 1.43–26.71). In various co-infection cases, the co-infection with two enteric pathogens was more commonly detected in diarrhea patients than non-diarrhea subjects (Table 6, p = 0.0079, OR = 5.86, 95% CI 1.35–25.41). However, the prevalence of co-infection with more than three enteric pathogens in patents was as much as in healthy controls.
Table 6
The co-infection of enteric pathogens detected in diarrhea cases and healthy controls
Co-infections of enteric pathogens
Diarrhea cases
n = 271
n (%)
Healthy controls
n = 149
n (%)
p value
OR (95% CI)
Any two any enteric pathogens
20 (7.4)
2 (1.3)
p = 0.0079
5.86 (1.35–25.41)
Virus–virus
9 (3.3)
0 (0.0)
p = 0.0298
 RVA–NoV GII
8 (3.0)
0 (0.0)
p = 0.0549
Bacteria–virus
5 (1.8)
0 (0.0)
p = 0.166
 DEC–NoV GII
4 (1.5)
0 (0.0)
p = 0.302
 DEC–RVA
3 (1.1)
0 (0.0)
p = 0.556
Bacteria–protozoan
3 (1.1)
2 (1.3)
p = 0.999
0.83 (0.14–5.00)
 DEC–B. hominis
3 (1.1)
2 (1.3)
p = 0.999
0.83 (0.14–5.00)
Any three enteric pathogens
1 (0.4)
0 (0.0)
p = 0.999
 DEC–RVA–Cryptosporidium spp.
1 (0.4)
0 (0.0)
p = 0.999
Total
21 (7.7)
2 (1.3)
p = 0.0059
6.17 (1.43–26.71)
Only co-infections with two pathogens found in at least 1% of diarrhea cases have been shown. The “–” symbol indicates the data can not be calculated
20 diarrhea cases of co-infections with two pathogens was identified, whereby two pathogens were identified, the prominent prevalence was virus–virus (45.0%, 9/20), followed by bacteria–virus (25.0%, 5/20) and bacteria–protozoan (15.0%, 3/20), and the other comprised co-infection was less common in diarrhea cases. The highest prevalence of co-infection in diarrhea cases was RVA–NoV GII (3.0%, n = 8), followed by DEC–NoV GII (1.5%, n = 4), DEC–RVA (1.1%, n = 3) and DEC–B. hominis (1.1%, n = 3). The prevalence of other co-infection between two pathogens was less than 1.0% in acute diarrhea cases (Table 6).

Discussion

Since most studies had focused on diarrheal illness in children under 5 years [6, 11], little is known about the prevalence of acute diarrhea caused by enteric pathogens among person over 5 years. This study was the first of its kind conducted to determine the enteropathogens of acute diarrheal disease in Yunnan Province, China, and a series of pathogens involving bacteria, viruses and parasites were examined with a combination of conventional and molecular diagnostic techniques.
The detection rate of at least one enteric pathogen was significantly higher in diarrhea cases than in healthy controls, which showed a wide range of pathogens involving bacteria, and similar results have also been obtained from other countries [29, 30]. Although bacteria and parasites were the prominent enteropathogen in acute diarrheal cases aged more than 5 years in some developing countries [31], to our surprise, viral pathogens (RVA and NoV) were the most common pathogen in present study.
DEC were detected with a PCR method in stool sample from the patients and non-diarrheal controls, and the result showed that DEC wasn’t the causative agent of diarrhea in individuals over 5 years, and similar conclusions were shown in another study [32]. However, the authors of the other study argued that DEC was one of important enteric pathogen causing acute diarrhea [33]. The detection rate of DEC in present study was lower than that presented in other study [32], but it was higher than that presented in other region of China [11]. The prevalence of DEC varies greatly in different regions due to the detection method [11], behavior habits, geography and environmental hygiene among different areas [34]. Although the molecular biology techniques (e.g. PCR and Real-time PCR) are useful for detecting DEC, PCR was not used widely in medical facilities because of constraints in many developing countries, including the poor laboratory conditions, limited funds and low detection capacities of staff [16]. Hence, DEC was not a pathogen that was routinely detected in clinical laboratories especially in low and middle income countries [35, 36]. The DEC was detected in many studies with the traditional serum agglutination method which has low sensitivity and specificity. Therefore, the prevalence of DEC was underestimated and the pathogenic spectrum of acute diarrheal illness was not accurately described [4]. It was accurately described to detect DEC by PCR with high sensitivity and specificity due to the following reasons [31]: Firstly, the clinical symptom of diarrhea caused by different DEC subtypes and other enteropathogens cannot be distinguished easily. Secondly, DEC is widely prevalent in food and environment, and the modern tourism and trade had accelerated the spread of DEC. The modern detection method (e.g. PCR) can improve the sensitivity and specificity for detecting DEC in stool samples in order to accurately assess the burden of DEC in cases [4, 31]. In addition, the modern method has advantages in saving diagnosis time and reducing workload of finding DEC in diarrhea cases. Especially, it is more accurate to identify the various DEC subtypes, and it can be completed more quickly and more accurately.
EAEC is also the leading cause of diarrhea in children, adult and HIV-positive patients worldwide [37, 38]. In addition, EAEC was one of major causes of diarrhea outbreak in some developed countries (e.g. Europe, the UK and Japan) [31, 38]. EAEC was not the important bacterial pathogen associated diarrhea in individuals over 5 years in present study, and similar conclusion was obtained from other study [32]. However another study showed that EAEC was associated with diarrheal disease [33]. Further studies found that the concentration with 1010 CFU of serotype 042 EAEC strain can lead to diarrhea, but other serotype of EAEC strain cannot cause diarrhea in children and adults [37, 38]. It can be deducted that the genotype is likely to be an important factor in determining pathogenicity. The detection rate of EAEC in this study was as high as 7.4%, which was similar to the other study [33]. However the prevalence of EAEC was still lower than in many developing countries [37]. In the present study, EPEC was also not associated with diarrhea disease, similar to other study [31]. Further mechanism research might be conducted to explore the pathogenicity and infectivity at a genetic level.
Adults suffering diarrhea rarely visit a medical institution, unless they have acute serious or persistent diarrhea. The study suggests that although many enteric pathogens were detected from diarrhea patients over 5 years old, only RVA was significantly related with diarrheal illness in individuals over 5 years old. This study provides further evidence that RVA is a cause of acute adult diarrhea in China, but other study show that RVA was not an etiological agent with diarrhea [32]. The frequency of RVA infection (7.4%) was close to other study (9.6%) [39], but was higher than in the study (2.6%) conducted in adolescents or adults (10–89 years) in Italy [40].
NoV GII is one of major pathogens which can lead sporadic and outbreak acute diarrhea cases across all age groups worldwide [41]. The present study showed that NoV GII was the second most common enteropathogen in diarrhea cases. The high prevalence of NoV GII in individuals might be attributed to frequent social activities, and NoV GII is one of the most important food borne pathogen and exists widely in foods (such as shellfish, vegetables and water, et al.). These foods contaminated with NoV GII were primary reasons to lead sporadic and outbreak acute diarrhea [4245]. The detection rate of NoV GII in our study was lower than that of in other study [46, 47]. The reason might be that seafood (e.g. shellfish) was not easily obtained and was not a conventional food in inland of China, including Kunming city.
Blastocystis hominis was found to be the most common protozoan in gastrointestinal tract of human and animals. It was widespread in natural world [48] and was highly prevalent in immunodeficiency patients [49]. Blastocystis hominis was not a pathogenic agent in present study, but other studies showed that B. hominis was a diarrhea-associated pathogen [50, 51]. Blastocystis hominis had high prevalence in healthy controls in present study implied that B. hominis was carried in health individual, which was a common phenomenon [50]. Whether B. hominis was one of pathogenic pathogen is need to explore the pathogenicity of different subtypes and mechanism. Cryptosporidium spp. and G. lamblia are leading cause of acute and chronic diarrhea in the tropics regions and some developing counties [52], but Cryptosporidium spp. had low prevalence and no one G. lamblia was detected in cases and healthy controls in present study, which indicating that these two kinds of intestinal protozoa were not serious disease burden and intimidate to individuals over 5 years old. This low prevalence of two protozoa might be due to epidemic characteristics of enteric parasites. Our research field was selected in urban with perfective municipal facilities of sewage treatment system, chlorine disinfection water, as well as, the population with high living level and health habits, so that the detection rate of enteric protozoa was very low, and the same results was showed in other studies in China [5, 17].
The co-infection was not neglected in diarrhea cases (7.7%) in this study, although other studies found that co-infection was high prevalent in sick individuals (13.0, 35.0, 25.0%, respectively) [12, 13, 53]. The co-infection leads to that individuals with greater levels of morbidity and mortality, making persons more vulnerable to species, for instance, the co-infection of RVA and other enteric pathogen can aggravate diarrheal symptom [14, 54]. In addition, the co-infection adds the difficulty to accurately determine etiological role of the enteric pathogen. Although co-infection by multiple groups of pathogens is the norm rather than the exception in nature, most research on the effect of pathogens on their hosts has been largely based on a single or few pathogen species [15]. Understanding the causes and consequences of co-infection among enteric pathogens remains one of the major challenges. Nevertheless, there is an increasing interest to move from the ‘diarrheal disease-one enteric pathogen’ perspective to a more holistic view of hosts as ecosystems of diarrhea illness [6], partially motivated by the health impact of co-occurring infections. In fact, in such complex ‘host–enteric pathogen ecosystems’ a variety of both direct and indirect interactions between enteric pathogens, their hosts and the circumstances must be taken into account [55].

Limitations of this study

It was indentified several limitations in this study. Firstly, the study was conducted in an urban region that probably shows a poor representation of the potential enteric pathogen. Secondly, the diarrhea cases were selected from outpatients and hospitalized cases. But the patients who did not to seek medical advice were not recruited. Thirdly, helminthes and some intestinal bacteria were not detected in this study. Fourthly, the percentage of diarrheal patients who have taken antibiotics before the admission was not known, which may influence the detection rate of bacterial pathogens. In addition, enteric protozoa were not detected with microscopy, and the concentration of DNA in 1 μL can be different and therefore, the outcome of PCR might not be comparable [56]. Therefore, further research involved diarrhea case from urban, rural, outpatient and hospitalized might be done to evaluate the burden of diarrhea disease and assess the association between diarrhea and specific enteric pathogen. Match case–control study will be a good choice, and quantitate the DNA by nanodrop or something else and then loaded equal amount of DNA (e.g. 1 ng) for every PCR reaction will be have high reliability for entire project.

Conclusions

Although it appears clear that RVA has impact on diarrhea illness, it was ignored in individuals over 5 years old. The prevalence of DEC was high in diarrhea cases, but it would be largely neglected due to lack of access to good quality diagnostic tests, which suggests that enhance laboratory capacities are urgently need in order to implement diarrhea surveillance programs. The co-infection was high prevalent in diarrhea cases, which will respond to better medical and public health interventions of diarrhea disease. In view of the diarrhea cases detected in urban region of Kunming city, Yunnan Province, which have effluent sewerage system, good sanitary condition and clean drinking water, it is concluded that food pollution might be the leading cause of acute gastroenteritis.

Authors’ contributions

SX-Z and ES performed the statistical analysis and drafted the manuscript. LG-T, JX-C, WH and XN-Z conceived and designed the study, CL-Y, LA, WP-G, XZ, SZ-L, SL, ZS-D and JH-C conducted the dates collected and pathogens detected, PY edited the English. All authors read and approved the final manuscript.

Acknowledgements

None.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Data of the study can be available upon request from the corresponding author (XN-Z).
The study was approved by the ethical review committee of the National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention. Informed verbal or written consent was obtained from the subject or their parents/guardians before collecting the stool samples.

Funding

Development of PCR to detect virus, G. lamblia was supported by the National Science and Technology Major Project (Grant number: 2008ZX10004-011); Development of PCR to find Cryptosporidium spp. was supported by National Science and Technology Major Project (2012ZX10004-220). The B. hominis identified was supported from National Natural Science Foundation of China (Grant number: 81473022). The field epidemiological investigation was supported by Chinese Special Program for Scientific Research of Public Health (No. 201502021). The bacterial pathogens identified was supported by The Fourth Round Three Year Action Plan Public Health of Shanghai city (GWIV-29). The data analysis was supported from the fund of the postdoctoral programme of the Fundação para a Ciência ea Tecnologia the Fundação para a Ciência ea Tecnologia, Portugal (SFRH/BPD/96637/2013).
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.
Literatur
1.
Zurück zum Zitat Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, Aggarwal R, Ahn SY, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the global burden of disease study 2010. Lancet. 2012;380(9859):2095–128. doi:10.1016/S0140-6736(12)61728-0.CrossRefPubMed Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, Aggarwal R, Ahn SY, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the global burden of disease study 2010. Lancet. 2012;380(9859):2095–128. doi:10.​1016/​S0140-6736(12)61728-0.CrossRefPubMed
3.
Zurück zum Zitat Platts-Mills JA, Babji S, Bodhidatta L, Gratz J, Haque R, Havt A, McCormick BJ, McGrath M, Olortegui MP, Samie A, et al. Pathogen-specific burdens of community diarrhoea in developing countries: a multisite birth cohort study (MAL-ED). Lancet Glob Health. 2015;3(9):e564–75. doi:10.1016/S2214-109X(15)00151-5.CrossRefPubMed Platts-Mills JA, Babji S, Bodhidatta L, Gratz J, Haque R, Havt A, McCormick BJ, McGrath M, Olortegui MP, Samie A, et al. Pathogen-specific burdens of community diarrhoea in developing countries: a multisite birth cohort study (MAL-ED). Lancet Glob Health. 2015;3(9):e564–75. doi:10.​1016/​S2214-109X(15)00151-5.CrossRefPubMed
4.
Zurück zum Zitat Liu J, Kabir F, Manneh J, Lertsethtakarn P, Begum S, Gratz J, Becker SM, Operario DJ, Taniuchi M, Janaki L, et al. Development and assessment of molecular diagnostic tests for 15 enteropathogens causing childhood diarrhoea: a multicentre study. Lancet Infect Dis. 2014;14(8):716–24. doi:10.1016/S1473-3099(14)70808-4.CrossRefPubMed Liu J, Kabir F, Manneh J, Lertsethtakarn P, Begum S, Gratz J, Becker SM, Operario DJ, Taniuchi M, Janaki L, et al. Development and assessment of molecular diagnostic tests for 15 enteropathogens causing childhood diarrhoea: a multicentre study. Lancet Infect Dis. 2014;14(8):716–24. doi:10.​1016/​S1473-3099(14)70808-4.CrossRefPubMed
5.
Zurück zum Zitat Hao R, Li P, Wang Y, Qiu S, Wang L, Li Z, Xie J, Wu Z, Lin R, Liu N, et al. Diversity of pathogens responsible for acute diarrheal disease in China. Clin Infect Dis. 2013;57(12):1788–90. doi:10.1093/cid/cit572.CrossRefPubMed Hao R, Li P, Wang Y, Qiu S, Wang L, Li Z, Xie J, Wu Z, Lin R, Liu N, et al. Diversity of pathogens responsible for acute diarrheal disease in China. Clin Infect Dis. 2013;57(12):1788–90. doi:10.​1093/​cid/​cit572.CrossRefPubMed
7.
Zurück zum Zitat Nic Fhogartaigh C, Dance DAB. Bacterial gastroenteritis. Medicine. 2013;41(12):693–9.CrossRef Nic Fhogartaigh C, Dance DAB. Bacterial gastroenteritis. Medicine. 2013;41(12):693–9.CrossRef
9.
Zurück zum Zitat Duan ZJ, Liu N, Yang SH, Zhang J, Sun LW, Tang JY, Jin Y, Du ZQ, Xu J, Wu QB, et al. Hospital-based surveillance of rotavirus diarrhea in the People’s Republic of China, August 2003–July 2007. J Infect Dis. 2009;200(Suppl 1):S167–73. doi:10.1086/605039.CrossRefPubMed Duan ZJ, Liu N, Yang SH, Zhang J, Sun LW, Tang JY, Jin Y, Du ZQ, Xu J, Wu QB, et al. Hospital-based surveillance of rotavirus diarrhea in the People’s Republic of China, August 2003–July 2007. J Infect Dis. 2009;200(Suppl 1):S167–73. doi:10.​1086/​605039.CrossRefPubMed
12.
Zurück zum Zitat Nimri LF, Elnasser Z, Batchoun R. Polymicrobial infections in children with diarrhoea in a rural area of Jordan. FEMS Immunol Med Microbiol. 2004;42(2):255–9.CrossRefPubMed Nimri LF, Elnasser Z, Batchoun R. Polymicrobial infections in children with diarrhoea in a rural area of Jordan. FEMS Immunol Med Microbiol. 2004;42(2):255–9.CrossRefPubMed
13.
Zurück zum Zitat Vu NT, Le Van P, Le Huy C, Nguyen GK, Weintraub A. Etiology and epidemiology of diarrhea in children in Hanoi, Vietnam. Int J Infect Dis. 2006;10(4):298–308.CrossRef Vu NT, Le Van P, Le Huy C, Nguyen GK, Weintraub A. Etiology and epidemiology of diarrhea in children in Hanoi, Vietnam. Int J Infect Dis. 2006;10(4):298–308.CrossRef
14.
Zurück zum Zitat Bhavnani D, Goldstick JE, Cevallos W, Trueba G, Eisenberg JNS. Synergistic effects between rotavirus and coinfecting pathogens on diarrheal disease: evidence from a community-based study in Northwestern Ecuador. Am J Epidemiol. 2012;176(5):387–95. doi:10.1093/aje/kws220.CrossRefPubMedPubMedCentral Bhavnani D, Goldstick JE, Cevallos W, Trueba G, Eisenberg JNS. Synergistic effects between rotavirus and coinfecting pathogens on diarrheal disease: evidence from a community-based study in Northwestern Ecuador. Am J Epidemiol. 2012;176(5):387–95. doi:10.​1093/​aje/​kws220.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Zhang Y, Zhao Y, Ding K, Wang X, Chen X, Liu Y, Chen Y. Analysis of bacterial pathogens causing acute diarrhea on the basis of sentinel surveillance in Shanghai, China, 2006–2011. Jpn J Infect Dis. 2014;67(4):264–8.CrossRefPubMed Zhang Y, Zhao Y, Ding K, Wang X, Chen X, Liu Y, Chen Y. Analysis of bacterial pathogens causing acute diarrhea on the basis of sentinel surveillance in Shanghai, China, 2006–2011. Jpn J Infect Dis. 2014;67(4):264–8.CrossRefPubMed
20.
Zurück zum Zitat Deer DM, Lampel KA. Development of a multiplex real-time PCR assay with internal amplification control for the detection of Shigella species and enteroinvasive Escherichia coli. J Food Prot. 2010;73(9):1618–25.PubMed Deer DM, Lampel KA. Development of a multiplex real-time PCR assay with internal amplification control for the detection of Shigella species and enteroinvasive Escherichia coli. J Food Prot. 2010;73(9):1618–25.PubMed
22.
Zurück zum Zitat Iturriza GM, Wong C, Blome S, Desselberger U, Gray J. Molecular characterization of VP6 genes of human rotavirus isolates: correlation of genogroups with subgroups and evidence of independent segregation. J Virol. 2002;76(13):6596–601.CrossRef Iturriza GM, Wong C, Blome S, Desselberger U, Gray J. Molecular characterization of VP6 genes of human rotavirus isolates: correlation of genogroups with subgroups and evidence of independent segregation. J Virol. 2002;76(13):6596–601.CrossRef
23.
Zurück zum Zitat Zintz C, Bok K, Parada E, Barnes-Eley M, Berke T, Staat MA, Azimi P, Jiang X, Matson DO. Prevalence and genetic characterization of caliciviruses among children hospitalized for acute gastroenteritis in the United States. Infect Genet Evol. 2005;5(3):281–90.CrossRefPubMed Zintz C, Bok K, Parada E, Barnes-Eley M, Berke T, Staat MA, Azimi P, Jiang X, Matson DO. Prevalence and genetic characterization of caliciviruses among children hospitalized for acute gastroenteritis in the United States. Infect Genet Evol. 2005;5(3):281–90.CrossRefPubMed
24.
Zurück zum Zitat Yan H, Yagyu F, Okitsu S, Nishio O, Ushijima H. Detection of norovirus (GI, GII), sapovirus and astrovirus in fecal samples using reverse transcription single-round multiplex PCR. J Virol Methods. 2003;114(1):37–44.CrossRefPubMed Yan H, Yagyu F, Okitsu S, Nishio O, Ushijima H. Detection of norovirus (GI, GII), sapovirus and astrovirus in fecal samples using reverse transcription single-round multiplex PCR. J Virol Methods. 2003;114(1):37–44.CrossRefPubMed
26.
Zurück zum Zitat Menounos PG, Spanakos G, Tegos N, Vassalos CM, Papadopoulou C, Vakalis NC. Direct detection of Blastocystis sp. in human faecal samples and subtype assignment using single strand conformational polymorphism and sequencing. Mol Cell Probes. 2008;22(1):24–9.CrossRefPubMed Menounos PG, Spanakos G, Tegos N, Vassalos CM, Papadopoulou C, Vakalis NC. Direct detection of Blastocystis sp. in human faecal samples and subtype assignment using single strand conformational polymorphism and sequencing. Mol Cell Probes. 2008;22(1):24–9.CrossRefPubMed
28.
Zurück zum Zitat Sulaiman IM, Fayer R, Bern C, Gilman RH, Trout JM, Schantz PM, Das P, Lal AA, Xiao L. Triosephosphate isomerase gene characterization and potential zoonotic transmission of Giardia duodenalis. Emerg Infect Dis. 2003;9(11):1444–52.CrossRefPubMedPubMedCentral Sulaiman IM, Fayer R, Bern C, Gilman RH, Trout JM, Schantz PM, Das P, Lal AA, Xiao L. Triosephosphate isomerase gene characterization and potential zoonotic transmission of Giardia duodenalis. Emerg Infect Dis. 2003;9(11):1444–52.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Albert MJ, Faruque AS, Faruque SM, Sack RB, Mahalanabis D. Case–control study of enteropathogens associated with childhood diarrhea in Dhaka, Bangladesh. J Clin Microbiol. 1999;37(11):3458–64.PubMedPubMedCentral Albert MJ, Faruque AS, Faruque SM, Sack RB, Mahalanabis D. Case–control study of enteropathogens associated with childhood diarrhea in Dhaka, Bangladesh. J Clin Microbiol. 1999;37(11):3458–64.PubMedPubMedCentral
30.
Zurück zum Zitat Randremanana R, Randrianirina F, Gousseff M, Dubois N, Razafindratsimandresy R, Hariniana ER, Garin B, Randriamanantena A, Rakotonirina HC, Ramparany L, et al. Case–control study of the etiology of infant diarrheal disease in 14 districts in Madagascar. PLoS ONE. 2012;7(9):e44533. doi:10.1371/journal.pone.0044533.CrossRefPubMedPubMedCentral Randremanana R, Randrianirina F, Gousseff M, Dubois N, Razafindratsimandresy R, Hariniana ER, Garin B, Randriamanantena A, Rakotonirina HC, Ramparany L, et al. Case–control study of the etiology of infant diarrheal disease in 14 districts in Madagascar. PLoS ONE. 2012;7(9):e44533. doi:10.​1371/​journal.​pone.​0044533.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Thompson CN, Phan MV, Hoang NV, Minh PV, Vinh NT, Thuy CT, Nga TT, Rabaa MA, Duy PT, Dung TT, et al. A prospective multi-center observational study of children hospitalized with diarrhea in Ho Chi Minh City, Vietnam. Am J Trop Med Hyg. 2015;92(5):1045–52. doi:10.4269/ajtmh.14-0655.CrossRefPubMedPubMedCentral Thompson CN, Phan MV, Hoang NV, Minh PV, Vinh NT, Thuy CT, Nga TT, Rabaa MA, Duy PT, Dung TT, et al. A prospective multi-center observational study of children hospitalized with diarrhea in Ho Chi Minh City, Vietnam. Am J Trop Med Hyg. 2015;92(5):1045–52. doi:10.​4269/​ajtmh.​14-0655.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Al-Gallas N, Bahri O, Bouratbeen A, Ben Haasen A, Ben Aissa R. Etiology of acute diarrhea in children and adults in Tunis, Tunisia, with emphasis on diarrheagenic Escherichia coli: prevalence, phenotyping, and molecular epidemiology. Am J Trop Med Hyg. 2007;77(3):571–82.PubMed Al-Gallas N, Bahri O, Bouratbeen A, Ben Haasen A, Ben Aissa R. Etiology of acute diarrhea in children and adults in Tunis, Tunisia, with emphasis on diarrheagenic Escherichia coli: prevalence, phenotyping, and molecular epidemiology. Am J Trop Med Hyg. 2007;77(3):571–82.PubMed
33.
Zurück zum Zitat Bruijnesteijn VCL, Dullaert-de BM, Ruijs GJ, van der Reijden WA, van der Zanden AG, Weel JF, Schuurs TA. Case–control comparison of bacterial and protozoan microorganisms associated with gastroenteritis: application of molecular detection. Clin Microbiol Infect. 2015;21(6):592-e9–19. doi:10.1016/j.cmi.2015.02.007.CrossRef Bruijnesteijn VCL, Dullaert-de BM, Ruijs GJ, van der Reijden WA, van der Zanden AG, Weel JF, Schuurs TA. Case–control comparison of bacterial and protozoan microorganisms associated with gastroenteritis: application of molecular detection. Clin Microbiol Infect. 2015;21(6):592-e9–19. doi:10.​1016/​j.​cmi.​2015.​02.​007.CrossRef
34.
Zurück zum Zitat Okeke IN. Diarrheagenic Escherichia coli in sub-Saharan Africa: status, uncertainties and necessities. J Infect Dev Ctries. 2009;3(11):817–42.PubMed Okeke IN. Diarrheagenic Escherichia coli in sub-Saharan Africa: status, uncertainties and necessities. J Infect Dev Ctries. 2009;3(11):817–42.PubMed
35.
Zurück zum Zitat Gerner-Smidt P, Jensen C, Olsen KE, Scheutz F, Molbak K, Olesen B. Diarrheagenic potential of Escherichia coli in children in a developed country. J Clin Microbiol. 2003;41(12):5836.CrossRefPubMedPubMedCentral Gerner-Smidt P, Jensen C, Olsen KE, Scheutz F, Molbak K, Olesen B. Diarrheagenic potential of Escherichia coli in children in a developed country. J Clin Microbiol. 2003;41(12):5836.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Estrada-Garcia T, Cerna JF, Paheco-Gil L, Velazquez RF, Ochoa TJ, Torres J, DuPont HL. Drug-resistant diarrheogenic Escherichia coli, Mexico. Emerg Infect Dis. 2005;11(8):1306–8.CrossRefPubMedPubMedCentral Estrada-Garcia T, Cerna JF, Paheco-Gil L, Velazquez RF, Ochoa TJ, Torres J, DuPont HL. Drug-resistant diarrheogenic Escherichia coli, Mexico. Emerg Infect Dis. 2005;11(8):1306–8.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Huang DB, Mohanty A, DuPont HL, Okhuysen PC, Chiang T. A review of an emerging enteric pathogen: enteroaggregative Escherichia coli. J Med Microbiol. 2006;55(Pt 10):1303–11.CrossRefPubMed Huang DB, Mohanty A, DuPont HL, Okhuysen PC, Chiang T. A review of an emerging enteric pathogen: enteroaggregative Escherichia coli. J Med Microbiol. 2006;55(Pt 10):1303–11.CrossRefPubMed
38.
Zurück zum Zitat Hebbelstrup JB, Olsen KE, Struve C, Krogfelt KA, Petersen AM. Epidemiology and clinical manifestations of enteroaggregative Escherichia coli. Clin Microbiol Rev. 2014;27(3):614–30. doi:10.1128/CMR.00112-13.CrossRef Hebbelstrup JB, Olsen KE, Struve C, Krogfelt KA, Petersen AM. Epidemiology and clinical manifestations of enteroaggregative Escherichia coli. Clin Microbiol Rev. 2014;27(3):614–30. doi:10.​1128/​CMR.​00112-13.CrossRef
40.
Zurück zum Zitat Ianiro G, Delogu R, Bonomo P, Fiore L, Ruggeri FM. Molecular analysis of group A rotaviruses detected in adults and adolescents with severe acute gastroenteritis in Italy in 2012. J Med Virol. 2014;86(6):1073–82. doi:10.1002/jmv.23871.CrossRefPubMed Ianiro G, Delogu R, Bonomo P, Fiore L, Ruggeri FM. Molecular analysis of group A rotaviruses detected in adults and adolescents with severe acute gastroenteritis in Italy in 2012. J Med Virol. 2014;86(6):1073–82. doi:10.​1002/​jmv.​23871.CrossRefPubMed
41.
Zurück zum Zitat Ahmed SM, Hall AJ, Robinson AE, Verhoef L, Premkumar P, Parashar UD, Koopmans M, Lopman BA. Global prevalence of norovirus in cases of gastroenteritis: a systematic review and meta-analysis. Lancet Infect Dis. 2014;14(8):725–30. doi:10.1016/S1473-3099(14)70767-4.CrossRefPubMed Ahmed SM, Hall AJ, Robinson AE, Verhoef L, Premkumar P, Parashar UD, Koopmans M, Lopman BA. Global prevalence of norovirus in cases of gastroenteritis: a systematic review and meta-analysis. Lancet Infect Dis. 2014;14(8):725–30. doi:10.​1016/​S1473-3099(14)70767-4.CrossRefPubMed
42.
44.
Zurück zum Zitat Crim SM, Iwamoto M, Huang JY, Griffin PM, Gilliss D, Cronquist AB, Cartter M, Tobin-D’Angelo M, Blythe D, Smith K, et al. Incidence and trends of infection with pathogens transmitted commonly through food—foodborne diseases active surveillance network, 10 US sites, 2006–2013. MMWR Morb Mortal Wkly Rep. 2014;63(15):328–32.PubMed Crim SM, Iwamoto M, Huang JY, Griffin PM, Gilliss D, Cronquist AB, Cartter M, Tobin-D’Angelo M, Blythe D, Smith K, et al. Incidence and trends of infection with pathogens transmitted commonly through food—foodborne diseases active surveillance network, 10 US sites, 2006–2013. MMWR Morb Mortal Wkly Rep. 2014;63(15):328–32.PubMed
46.
Zurück zum Zitat Tian G, Jin M, Li H, Li Q, Wang J, Duan ZJ. Clinical characteristics and genetic diversity of noroviruses in adults with acute gastroenteritis in Beijing, China in 2008–2009. J Med Virol. 2014;86(7):1235–42. doi:10.1002/jmv.23802.CrossRefPubMed Tian G, Jin M, Li H, Li Q, Wang J, Duan ZJ. Clinical characteristics and genetic diversity of noroviruses in adults with acute gastroenteritis in Beijing, China in 2008–2009. J Med Virol. 2014;86(7):1235–42. doi:10.​1002/​jmv.​23802.CrossRefPubMed
47.
Zurück zum Zitat Jin M, Chen J, Zhang XH, Zhang M, Li HY, Cheng WX, Liu N, Tan M, Jiang T, Duan ZJ. Genetic diversity of noroviruses in Chinese adults: potential recombination hotspots and GII-4/Den Haag-specific mutations at a putative epitope. Infect Genet Evol. 2011;11(7):1716–26. doi:10.1016/j.meegid.2011.07.005.CrossRefPubMed Jin M, Chen J, Zhang XH, Zhang M, Li HY, Cheng WX, Liu N, Tan M, Jiang T, Duan ZJ. Genetic diversity of noroviruses in Chinese adults: potential recombination hotspots and GII-4/Den Haag-specific mutations at a putative epitope. Infect Genet Evol. 2011;11(7):1716–26. doi:10.​1016/​j.​meegid.​2011.​07.​005.CrossRefPubMed
48.
Zurück zum Zitat Thathaisong U, Worapong J, Mungthin M, Tan-Ariya P, Viputtigul K, Sudatis A, Noonai A, Leelayoova S. Blastocystis isolates from a pig and a horse are closely related to Blastocystis hominis. J Clin Microbiol. 2003;41(3):967–75.CrossRefPubMedPubMedCentral Thathaisong U, Worapong J, Mungthin M, Tan-Ariya P, Viputtigul K, Sudatis A, Noonai A, Leelayoova S. Blastocystis isolates from a pig and a horse are closely related to Blastocystis hominis. J Clin Microbiol. 2003;41(3):967–75.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Wang KX, Li CP, Wang J, Cui YB. Epidemiological survey of Blastocystis hominis in Huainan City, Anhui Province, China. World J Gastroenterol. 2002;8(5):928–32.CrossRefPubMedPubMedCentral Wang KX, Li CP, Wang J, Cui YB. Epidemiological survey of Blastocystis hominis in Huainan City, Anhui Province, China. World J Gastroenterol. 2002;8(5):928–32.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Kotloff KL, Nataro JP, Blackwelder WC, Nasrin D, Farag TH, Panchalingam S, Wu Y, Sow SO, Sur D, Breiman RF, et al. Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicenter Study, GEMS): a prospective, case–control study. Lancet. 2013;382(9888):209–22. doi:10.1016/S0140-6736(13)60844-2.CrossRefPubMed Kotloff KL, Nataro JP, Blackwelder WC, Nasrin D, Farag TH, Panchalingam S, Wu Y, Sow SO, Sur D, Breiman RF, et al. Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicenter Study, GEMS): a prospective, case–control study. Lancet. 2013;382(9888):209–22. doi:10.​1016/​S0140-6736(13)60844-2.CrossRefPubMed
53.
Zurück zum Zitat Li LL, Liu N, Humphries EM, Yu JM, Li S, Lindsay BR, Stine OC, Duan ZJ. Aetiology of diarrhoeal disease and evaluation of viral-bacterial coinfection in children under 5 years old in China: a matched case–control study. Clin Microbiol Infect. 2016;22(4):381.e9–16. doi:10.1016/j.cmi.2015.12.018.CrossRef Li LL, Liu N, Humphries EM, Yu JM, Li S, Lindsay BR, Stine OC, Duan ZJ. Aetiology of diarrhoeal disease and evaluation of viral-bacterial coinfection in children under 5 years old in China: a matched case–control study. Clin Microbiol Infect. 2016;22(4):381.e9–16. doi:10.​1016/​j.​cmi.​2015.​12.​018.CrossRef
54.
Zurück zum Zitat Valentini D, Vittucci AC, Grandin A, Tozzi AE, Russo C, Onori M, Menichella D, Bartuli A, Villani A. Coinfection in acute gastroenteritis predicts a more severe clinical course in children. Eur J Clin Microbiol Infect Dis. 2013;32(7):909–15. doi:10.1007/s10096-013-1825-9.CrossRefPubMed Valentini D, Vittucci AC, Grandin A, Tozzi AE, Russo C, Onori M, Menichella D, Bartuli A, Villani A. Coinfection in acute gastroenteritis predicts a more severe clinical course in children. Eur J Clin Microbiol Infect Dis. 2013;32(7):909–15. doi:10.​1007/​s10096-013-1825-9.CrossRefPubMed
55.
Zurück zum Zitat Seabloom EW, Borer ET, Gross K, Kendig AE, Lacroix C, Mitchell CE, Mordecai EA, Power AG. The community ecology of pathogens: coinfection, coexistence and community composition. Ecol Lett. 2015;18(4):401–15. doi:10.1111/ele.12418.CrossRefPubMed Seabloom EW, Borer ET, Gross K, Kendig AE, Lacroix C, Mitchell CE, Mordecai EA, Power AG. The community ecology of pathogens: coinfection, coexistence and community composition. Ecol Lett. 2015;18(4):401–15. doi:10.​1111/​ele.​12418.CrossRefPubMed
Metadaten
Titel
Case–control study of diarrheal disease etiology in individuals over 5 years in southwest China
verfasst von
Shun-Xian Zhang
Chun-Li Yang
Wen-Peng Gu
Lin Ai
Emmanuel Serrano
Pin Yang
Xia Zhou
Shi-Zhu Li
Shan Lv
Zhi-Sheng Dang
Jun-Hu Chen
Wei Hu
Li-Guang Tian
Jia-Xu Chen
Xiao-Nong Zhou
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Gut Pathogens / Ausgabe 1/2016
Elektronische ISSN: 1757-4749
DOI
https://doi.org/10.1186/s13099-016-0141-1

Weitere Artikel der Ausgabe 1/2016

Gut Pathogens 1/2016 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.