Skip to main content

Changes in fecal microbiota of dairy cows with and without endometritis

Abstract

Background

Endometritis is a uterine infection caused by bacterial pathogens and has detrimental effects on productive and reproductive performance in dairy cows. A large number of studies have demonstrated the association of gut microbiota with infectious diseases. However, the role of gut microbiota in dairy cows with endometritis is still poorly understood.

Results

In the present study, we characterized the fecal microbial populations in the dairy cows suffering from metritis (n = 10) and healthy cows (n = 9) using the 16 S rRNA gene sequencing. Results revealed an increased abundance of Firmicutes and Bacteroidetes in the affected cows indicating the potential role of these two bacterial taxa in the pathogenesis of endometritis. The Ruminococcaceae_UCG-005 was the predominant genus while Olsenella and Succinivibrio were the most abundant genera in the cows affected with metritis. Further, the association of specific genera from Firmicutes and Bacteroidetes indicated three co-occurrence groups indicating the potential interaction of these genera in modulating the immune response, dysbiosis and inflammatory reaction. In addition, a significantly higher abundance of genes involved in the excretory system was observed in affected cows.

Conclusions

Our findings provide evidence of changes in gut microbiota composition in cows suffering from metritis and advocate the need to explore the effect of commensal gut bacteria specifically co-occurring taxa in uterine inflammation and infection.

Peer Review reports

Background

Endometritis is a big concern for the dairy industry worldwide because it causes huge economic losses due to reduced milk production, lower conception rates, early culling, increased use of antibiotics, and treatment costs [1,2,3]. For example, the annual economic losses from cow uterine infections amounted to $1.4 billion in Europe and $650 million in the United States [4]. Bacterial contamination of the uterine lumen following parturition has long been documented as the main cause of endometritis. To date, various pathogens, including Escherichia coli, Arcanobacterium pyogenes, Fusobacterium necrophorum, Prevotella melaninogenicus, Bacteroidetes spp., Pseudomonas spp., Streptococcus spp., and Staphylococcus spp., have been identified in a variety of combinations from cows diagnosed with postpartum metritis to support the bacterial hypothesis of the pathogenesis of these uterine infections [5,6,7].

A better understanding of the microbial communities in the gastrointestinal tract is critical for developing efficient therapeutic interventions for endometritis. It is well established that gut microbiota plays a key role in the maintenance of health and development of disease [8,9,10]. To date, a large number of studies have reported the composition and function of the uterine bacterial community in animals, such as cattle [11], sheep [12], and humans [13]. Even though the onset of many infectious diseases including endometritis is known to be associated with gut microbiota, very few studies are available on this aspect in dairy cows. Therefore, it is imperative to understand the association of the gut microbiome with endometritis in dairy heifers.

With the advent and widespread use of next-generation sequencing technologies, conducting deep sequencing on samples from specific environments became feasible and allows investigation of the relationship between microbial community and disease. This strategy has been widely used in many animals, such as cattle [14], sheep [15], goat [16], buffalo [17], and humans [18]. Moreover, it also allows the study of the association between gut bacterial community and endometritis by 16 S rDNA sequencing is feasible. The present study aimed to characterize the gut microbiota and its functional diversity in cows affected with metritis.

Results

Sequence information

The sequence statistics of the 16 S rRNA gene for 19 samples are listed in Table 1. In total, 2,685,560 raw sequences were generated from all the samples, with a mean number of 141,345.26 raw tags for each sample. After filtering, a total of 2,562,823 clean tags were obtained, indicating that a total of approximately 95.54% clean raw data was available. Most clean tags were mainly distributed in the range of 400–440 bp (Fig. 1A). Cluster analysis of clean reads yielded a total of 2,620 OTUs, of which 2,174 OTUs were common between the metritis and healthy groups (Fig. 1B). In addition, a total of 195 and 251 OTUs were specific to metritis and healthy groups, respectively. Moreover, we identified a total of 311 core OTUs shared among the samples (Fig. 1C).

Table 1 Statistics information of 16 S rRNA sequencing for 19 samples
Fig. 1
figure 1

Distribution of clean tags (A) and Venn analysis of OTUs between groups (B) or samples (C)

Fecal bacterial community structure and composition

To compare the fecal bacterial composition of healthy and affected cows, we performed the 16 S rRNA gene amplicon sequencing. The alpha diversity analysis was used to compare the richness and diversity of bacterial communities between healthy and affected cows. The results revealed the lower values (P < 0.05) of observed species, Chao1, and Shannon indices in the affected cows (Fig. 2A), indicating a lower gut bacterial richness. However, no difference in the Simpson index was observed between the two groups revealing comparable levels of species diversity (Fig. 2A). The rarefaction curve indicated that the sequencing data were reliable and the abundance of different taxa varied depending on the sample (Figure S1). Further, we determined the beta diversity using the partial least squares discrimination analysis which indicated that gut bacteria in affected cows clustered separately from healthy cows (Fig. 2B).

The OTUs were assigned to 15 phyla, 26 classes, 42 orders, 74 families, and 210 genera. At the phylum level, Firmicutes, Bacteroidetes, Actinobacteria, Tenericutes, Proteobacteria, Saccharibacteria, and Spirochaetae were the major taxa with the relative abundance of more than 0.5% of all bacteria; Firmicutes was the predominant phylum, representing more than 68% and 71% of total bacteria present in affected and healthy cows, respectively (Fig. 2C). At the genus level, a total of 27 bacterial genera were present in affected cows, while the healthy cows had 32 genera with an abundance of more than 0.5% (Table S1). Notably, Ruminococcaceae_UCG-005 was the predominant genus in the affected and healthy groups representing 18.51% and 15.83% of all bacteria, respectively (Fig. 2D).

Fig. 2
figure 2

Comparison of gut microbiota profiles between metritis and healthy cows. (A) Alpha diversity in each microbiota was compared between metritis and healthy cows. (B) Partial Least Squares Discrimination analysis of beta diversity between metritis and healthy cows. (C) Bar chart representing the microbiota compositions at the phylum level between metritis and healthy cows. (D) Bar chart representing the microbiota compositions at the genus level between metritis and healthy cows

To differentiate significant bacterial taxa between affected and healthy cows, Wilcoxon-test and LefSe analysis were used. The results revealed that the relative abundance of Olsenella, Succinivibrio, Atopobium, and Eubacterium oxidoreducens group was increased in affected cows, while the abundance of Treponema2, Ruminiclostridium9, Lachnospiraceae UCG001, Caproiciproducens, and Papillibacter was increased in the healthy group (Fig. 3A). The LefSe analysis indicated that the relative abundance of Olsenella, Lachnospiraceae NC2004, and Succinivibrio were increased in the affected cows, while the abundance of Lachnospiraceae FCS020, Treponema2, and Pseudoramibacter was higher in healthy cows (Fig. 3B). Interestingly, both methods confirmed that Olsenella and Succinivibrio had a higher relative abundance in the affected cows compared to healthy ones.

Fig. 3
figure 3

Differences analysis in the gut microbiota between metritis and health cows using the Wilcoxon test (A) and Lefse analysis(B)

Co-occurrence of bacterial genera

As shown in Fig. 4, we identified three major co-occurrence groups (COGs) of bacterial communities that included genera belonging to Firmicutes and Bacteriodetes. One COG group is constituted of three genera including Paeniclostridium, Clostridium_sensu_stricto_1, and Prevotellaceae_UCG_003. While second COG group consisted of four genera including Bacteroids, Alistipes, Rikenellaceae_RC9_gut_group, and Ruminococcaeae_UCG_010. The third COG group had four genera including Tyzzerella_4, Ruminococcaeae_UCG_005, Eubacterium coprostanoligenes_group and Christenallaceae_R7_group.

Fig. 4
figure 4

Co-occurrence network of genera observed in fecal microbiota

Functional prediction of gut bacterial communities

To further predict the function of bacteria genes in affected and healthy cows, the PICRUSt2 software was used. In total, the genes were annotated to 6 level-1 and 44 level-2 KEGG pathways (Table 2). The most abundant first-level pathways were metabolism and human diseases. Among them, the genes for carbohydrate metabolism were abundant in the metabolism pathway, while drug resistance (antimicrobial) had a higher relative abundance in the human diseases pathway. The analysis exhibited significant differences (P < 0.05) in the relative abundance of genes coding for Signal transduction, Replication and repair, and Excretory system pathways between affected and healthy cows. Moreover, using the STAMP analysis, carbohydrate metabolism and excretory system pathways were found to be significantly enriched (P < 0.05) between the affected and healthy groups (Fig. 5).

Table 2 Relative abundance analysis of the functional microbial genes in the metritis and health feces
Fig. 5
figure 5

Abundance profiles of predicted functional gene categories showed significant statistical differences between metritis and healthy cows. The results were filtered using a P-value of 0.05 and effective size of 0.05 threshold in STAMP

Discussion

Endometritis is highly prevalent in dairy cows and is characterized by the inflammation of endometrial glandular and stromal tissues. It has been reported that the pathogenesis of endometritis is complicated and many causative pathogens have been successively identified, such as the Escherichia coli, Trueperella pyogenes, Fusobacterium necrophorum, and Staphylococcus aureus [6]. To date, studies on endometritis pathogens have mainly concentrated on the composition and function of the uterine microbiota in cattle [1, 7, 11]. However, no studies have reported the potential role of changes in gut microbiota in cows suffering from metritis. To the best of our knowledge, this is the first report on the potential role of the gut bacteriome in the onset of metritis in cows compared with the healthy ones. In the present study, an increased abundance of Firmicutes and Bacteroidetes in cows affected with metritis. Similarly, both Firmicutes and Bacteroidetes were also found to be abundant in the uterine microbiota in cattle [1, 7, 11]. These results further suggested that gut bacteria might play vital roles in maintaining the homeostasis of the intrauterine environment. However, more work is needed to confirm the potential role of these microbes in the pathogenesis of metritis.

It is well established that changes in the gut microbiota are frequently associated with disease incidence [19,20,21]. Gut microbiota has an important impact on disease and health, and its mechanism mainly includes the following aspects: immune regulation, nutrient metabolism, inflammation regulation, toxin metabolism. Understanding the mechanisms of how gut microbiota affects disease and health could help us find more effective interventions to maintain the balance of gut microbiota and promote health. Unveiling the diversity of gut microbiota helps in mining the pathogens affecting the disease. In the present study, relatively higher abundance of Olsenella and Succinivibrio was observed in the affected cows compared to healthy group. Association of Olsenella with reduction in gut inflammation has been reported earlier [22], suggesting its potential role in inflammatory conditions and host health. Evidence showed that Succinivibrio strains are associated with the production of acetic and succinic acids [23]. Recently, studies have demonstrated that succinic acid was upregulated in cows with endometritis using the metabolomic analysis [24], which indicates that the Succinivibrio might play a vital role in regulating the host health.

The diversity of the gut microbiota is strongly associated with changes in gut microbial functions [25]. In the present study, the most abundant first-level pathways were Metabolism and Human Diseases in the affected and healthy cows. Further, we observed the excretory system pathway was found to be significantly different between affected and healthy groups using STAMP analysis (P < 0.05). Previous studies have demonstrated that the excretory system is responsible for not only regulating the water balance in various body fluids but also maintains the chemical composition of body fluids by removing metabolic wastes [26,27,28]. It can be inferred that gut microbiota function in the intrauterine environment through the excretory system, thereby affecting the host’s health.

The present study revealed four COGs of fecal bacteria. First COG exhibiting interaction of Clostridium genera with Prevotellacaea is in agreement with earlier reports indicating Clostridium is a major genus present in vaginal and fecal microbiota in cows [29, 30]. An increased abundance of Clostridium species in fecal samples at pre-breeding in animals that were unable to establish a pregnancy as this spp. can interact with other microbes to induce regulatory T cells or anti-inflammatory commensal bacteria leading to microbial dysbiosis in the vaginal ecosystem. Moreover, Prevotella spp. is reported as one of the most common microbes that cause uterine infections and also linked to bacterial vaginosis [31, 32]. So this correlation of Clostrium genera with Prevotella spp. seems meaningful regarding their potential involvement in uterine infections including endometritis [29, 33]. The second COG group observed in the present study consisted of four genera including Bacteroids, Alistipes, Rikenellaceae_RC9_gut_group, and Ruminococcaeae_UCG_010. These findings are in agreement with earlier studies reporting both Bacteroids and Ruminococcaeae positively correlated in a COG associated with endometritis in dairy cows [34, 35]. Moreover, the fecal abundance of Bacteroides, Clostridium and Alistipes species can be used as a marker for the prediction of chances to establish a pregnancy in a cow. Additionally, Bacteroides species are also associated with negative health status in cattle [36]. Likewise, increased abundance of Alistipes, Ruminococcaceae UCG-005, Ruminococcaceae UCG-013, and Prevotella in addition to other genera, was associated with CXCL13 which is a promising marker for chronic inflammation of the endometrium [37]. Moreover, the interaction of Bacteroides fragilis with Burkholderiales indicated potential immunomodulatory effects through pyrine-caspase1 inflammasome formation subsequently leading to activation of the TLR2/TLR4 signaling pathway [38].

The third COG was constituted of four bacterial genera including Tyzzerella_4, Ruminococcaeae_UCG_005, Eubacterium coprostanoligenes_group and Christenallaceae_R7_group. Negative association of Tyzzerella_4 with serum levels of progesterone and testosterone has been observed in human subjects with postpartum depressive disorder. It should be noted that high levels of estrogen have shown association with pathogenesis of endometriosis which is an estrogen-dependent disease [39]. Due to negative effect of Tyzzerella_4 with progesterone might lead to increased estrogen levels subsequently affecting onset of metritis.

Findings of the present study revealed that uterine pathogens might interact with each other in avoiding uterine defense and interact to facilitate colonization of the endometrium. Similar interactions of microbial pathogens were also observed in cows with metritis or purulent vaginal discharged [1, 40]. Pathogenic bacteria (such as Trueperella spp., Fusobacterium spp.) were also present in the uterus of virgin heifers and of pregnant cows [41, 42]. Collectively, the co-occurrence of uterine pathogens could be considered of major importance in the development of uterine infection. The cooperative interspecies signaling and mechanism behind synergisms need to be elucidated.

The favorable interactions among bacteria in COG including metabolite exchanges, could promote group survival under adverse conditions like nutrient deficiencies [43]. Variations in bacterial composition in the uterus of healthy and diseased cows indicates effect of nutritional and physiological changes. Identifying the co-occurring microbes and critical nutrients that favor growth of pathogenic bacteria is crucial for understanding the disease pathogenesis and developing novel treatment strategies [44]. Such competitive and cooperative interactions of gut microbes and mechanisms of establishing infection requires further elucidation through future studies involving metabolomic and transcriptomic data.

In this work, we characterized the changes in gut microbiota composition in cows suffering from endometritis in dairy cows. However, the study has limitations in terms of the causal relationship between the gut microbiome and endometritis. Further investigation is necessary to resolve the issue.

Conclusions

The findings of present study revealed the association between the gut bacteriome and endometritis in dairy cows. The Ruminococcaceae_UCG-005 was the predominant genus while Olsenella and Succinivibrio were most abundant genera in the cows affected with metritis. Three co-occurrences bacteria group identified in the present study reveal the role of genera from Firmicutes and Bacteroidetes phyla in involvement in metritis. Our findings indicated the potential association of functional genes related to excretory system pathways with endometritis in dairy cows. This study is of great significance for improving the level of animal reproductive health, and is helpful to provide more effective strategies and measures for the prevention and treatment of obstetric diseases.

Methods

Sample collection

The fecal samples were collected in Jan 2020 from 15 days postpartum Holstein cows (n = 19) housed in the experimental cattle farm of Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences. Incidence of metritis was determined according to method as described previously [45]. Briefly, cows showing clinical signs including red-brown to purulent foul-smelling vaginal discharge; transrectal palpation of a thin-walled, large uterus containing large amounts of fluid or gas, and absence of systemic clinical signs like pyrexia, anorexia or reduced milk production. Cows with none of the above-mentioned clinical signs (uterine size and thickness expected for 5–10 days in milk, a watery red vaginal discharge without foul odor, and no systemic clinical signs) were classified as healthy. No medication was administered prior to sample collection. Put on shoulder-length gloves, lubricate the anus with paraffin oil, grasp the tail with the other hand, raise the head, pick up the fingers together, form a 45–60 degree angle, into the rectum. Discard the first 3 handfuls of manure and collect the fourth handfuls to the RNA-free 50mL centrifuge tube. The samples were flash-frozen on dry ice and stored at − 80 °C until DNA extraction.

DNA extraction

DNA was isolated from each fecal sample using the QIAamp Microbiome Kit (Qiagen, Hilden, Germany) following the manufacturer’s instructions. Concentration and purity of metagenomic DNA were measured by NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Waltham, MA). The quality of metagenomic DNA was confirmed by 1% agarose gel electrophoresis.

16 S rRNA gene sequencing

The V3 and V4 regions of 16 S rRNA gene were amplified using the primer pairs described by [46]. The polymerase chain reaction (PCR) conditions for amplification were: denaturation at 94 °C for 3 min followed by 30 cycles of 45 s at 94 °C, 60 s at 52 °C, and 60 s at 72 °C, with a final elongation at 72 °C for 10 min. Amplified PCR products were evaluated by 1% agarose gel electrophoresis. The PCR products were purified with the Agencourt AMPure XP (Beckman Coulter, USA) following the manufacturer’s instructions. The Nextera XT Index kit (Illumina, San Diego, CA, USA) was used to construct the DNA library for each sample. Normalization was performed with a Nextera XT DNA library prep kit (Illumina, San Diego, CA, USA), and paired-end sequencing of each sample was performed through the Illumina MiSeq platform (Illumina, San Diego, CA, USA).

Bioinformatic Analysis

Raw sequence data for each sample was trimmed and filtered by using FLASH ver1.20 [47] and Trimmomatic ver0.36 [48] software. In addition, the sequences shorter than 120 base pairs were further discarded. Subsequently, the Quantitative Insights into Microbial Ecology (QIIME; version2) pipeline was used for filtering the low-quality tags while retaining effective tags [49]. The filtered reads were clustered as operational taxonomic unit (OTU) at 97% similarity using the Uparse ver. 7.0.1001 software [50]. OTUs were taxonomically assigned using the RDP Classifier algorithm against the Silva database (ver.132) with the confidence threshold of 0.7. A p-value < 0.05 was defined as the significant threshold level.

Statistical analyses

Microbial alpha diversity parameters including Chao1, Shannon, and Simpson were analyzed using the Mothur ver.1.48.0 software [51]. Partial Least Squares Discrimination analysis (PLS-DA) was used to assess microbial beta diversity at the OTU level. Three multivariate statistical tests (ANOSIM, MetaStats, and LefSe) were used to evaluate the differences in species composition and community structure between healthy and affected cows. The functional prediction of 16 S rRNA genes were performed by the PICRUSt2 software [52]. Amino acid sequences were aligned and translated from the gene catalog against the proteins in the KEGG database. The hierarchical Ward-linkage clustering was used to define genus COGs as described previously [53]. The R program and Tutools platform (https://www.cloudtutu.com/) were used for visualizing of the data and statistical analysis.

Data availability

The sequence data have been deposited at NCBI Sequence Read Archive database with accession number PRJNA847011. All data generated or analyzed during this study are included in this published article and its additional files, and the dataset analyzed during the current study is available from the corresponding author on reasonable request.

Abbreviations

COGs:

co-occurrence groups

KEGG:

Kyoto Encyclopedia of Genes and Genomes

LefSe:

Linear discriminant analysis Effect Size

OTU:

operational taxonomic unit

PCR:

polymerase chain reaction

PLS-DA:

Partial Least Squares Discrimination analysis

rRNA:

ribosomal Ribonucleic Acid

References

  1. Bicalho MLS, Machado VS, Higgins CH, Lima FS, Bicalho RC. Genetic and functional analysis of the bovine uterine microbiota. Part I: Metritis versus healthy cows. J Dairy Sci. 2017;100(5):3850–62.

    Article  CAS  PubMed  Google Scholar 

  2. Drillich M, Beetz O, Pfützner A, Sabin M, Sabin HJ, Kutzer P, Nattermann H, Heuwieser W. Evaluation of a systemic antibiotic treatment of toxic puerperal metritis in dairy cows. J Dairy Sci. 2001;84(9):2010–7.

    Article  CAS  PubMed  Google Scholar 

  3. Sheldon IM, Lewis GS, LeBlanc S, Gilbert RO. Defining postpartum uterine disease in cattle. Theriogenology. 2006;65(8):1516–30.

    Article  PubMed  Google Scholar 

  4. Wang X, Tian F, Chen C, Feng Y, Sheng X, Guo Y, Ni H. Exosome-derived uterine microRNAs isolated from cows with endometritis impede blastocyst development. Reprod Biol. 2019;19(2):204–9.

    Article  CAS  PubMed  Google Scholar 

  5. Azawi OI. Postpartum uterine infection in cattle. Anim Reprod Sci. 2008;105(3–4):187–208.

    Article  CAS  PubMed  Google Scholar 

  6. Galvão KN, Bicalho RC, Jeon SJ. Symposium review: The uterine microbiome associated with the development of uterine disease in dairy cows. J Dairy Sci 2019; 102(12):11786–11797.

  7. Pascottini OB, Van Schyndel SJ, Spricigo JFW, Rousseau J, Weese JS, LeBlanc SJ. Dynamics of uterine microbiota in postpartum dairy cows with clinical or subclinical endometritis. Sci Rep. 2020;10(1):12353.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Adak A, Khan MR. An insight into gut microbiota and its functionalities. Cell Mol Life Sci. 2019;76(3):473–93.

    Article  CAS  PubMed  Google Scholar 

  9. Arvonen M, Berntson L, Pokka T, Karttunen TJ, Vähäsalo P, Stoll ML. Gut microbiota-host interactions and juvenile idiopathic arthritis. Pediatr Rheumatol Online J. 2016;14(1):44.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Huipeng W, Lifeng G, Chuang G, Jiaying Z, Yuankun C. The differences in colonic mucosal microbiota between normal individual and colon cancer patients by polymerase chain reaction-denaturing gradient gel electrophoresis. J Clin Gastroenterol. 2014;48(2):138–44.

    Article  PubMed  Google Scholar 

  11. Santos TM, Gilbert RO, Bicalho RC. Metagenomic analysis of the uterine bacterial microbiota in healthy and metritic postpartum dairy cows. J Dairy Sci. 2011;94(1):291–302.

    Article  CAS  PubMed  Google Scholar 

  12. Han J, Wang M, Pan Y, Hu X, Zhang X, Cui Y, Xu G, Wang L, Yu S. [Establishment and evaluation of multiplex PCR for detection of main pathogenic bacteria of endometritis in tibetan sheep]. Sheng Wu Gong Cheng Xue Bao. 2020;36(5):908–19.

    CAS  PubMed  Google Scholar 

  13. Lozano FM, Bernabeu A, Lledo B, Morales R, Diaz M, Aranda FI, Llacer J, Bernabeu R. Characterization of the vaginal and endometrial microbiome in patients with chronic endometritis. Eur J Obstet Gynecol Reprod Biol. 2021;263:25–32.

    Article  CAS  PubMed  Google Scholar 

  14. Cendron F, Niero G, Carlino G, Penasa M, Cassandro M. Characterizing the fecal bacteria and archaea community of heifers and lactating cows through 16S rRNA next-generation sequencing. J Appl Genet. 2020;61(4):593–605.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Fan Q, Cui X, Wang Z, Chang S, Wanapat M, Yan T, Hou F. Rumen Microbiota of Tibetan Sheep (Ovis aries) adaptation to extremely cold season on the Qinghai-Tibetan Plateau. Front Vet Sci. 2021;8:673822.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Wang L, Wu D, Yan T, Wang L. The impact of rumen cannulation on the microbial community of goat rumens as measured using 16S rRNA high-throughput sequencing. J Anim Physiol Anim Nutr (Berl). 2018;102(1):175–83.

    Article  CAS  PubMed  Google Scholar 

  17. Marino M, Dubsky de Wittenau G, Saccà E, Cattonaro F, Spadotto A, Innocente N, Radovic S, Piasentier E, Marroni F. Metagenomic profiles of different types of Italian high-moisture mozzarella cheese. Food Microbiol. 2019;79:123–31.

    Article  CAS  PubMed  Google Scholar 

  18. Boers SA, Jansen R, Hays JP. Understanding and overcoming the pitfalls and biases of next-generation sequencing (NGS) methods for use in the routine clinical microbiological diagnostic laboratory. Eur J Clin Microbiol Infect Dis. 2019;38(6):1059–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Takagi T, Naito Y, Kashiwagi S, Uchiyama K, Mizushima K, Kamada K, Ishikawa T, Inoue R, Okuda K, Tsujimoto Y, et al. Changes in the Gut Microbiota are Associated with Hypertension, Hyperlipidemia, and type 2 diabetes Mellitus in Japanese subjects. Nutrients. 2020;12(10):2996.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Jackson MA, Verdi S, Maxan M-E, Shin CM, Zierer J, Bowyer RCE, Martin T, Williams FMK, Menni C, Bell JT, et al. Gut microbiota associations with common diseases and prescription medications in a population-based cohort. Nat Commun. 2018;9(1):2655.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Albhaisi SAM, Bajaj JS, Sanyal AJ. Role of gut microbiota in liver disease. Am J Physiol Gastrointest Liver Physiol. 2020;318(1):G84–98.

    Article  CAS  PubMed  Google Scholar 

  22. Methé BA, Nelson KE, Pop M, Creasy HH, Giglio MG, Huttenhower C, Gevers D, Petrosino JF, Abubucker S, Badger JH, et al. A framework for human microbiome research. Nature. 2012;486(7402):215–21.

    Article  PubMed Central  Google Scholar 

  23. Bryant MP, Small N. Characteristics of two new genera of anaerobic curved rods isolated from the rumen of cattle. J Bacteriol. 1956;72(1):22–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang Y, Chen T, Gan Z, Li H, Li Y, Zhang Y, Zhao X. Metabolomic analysis of untargeted bovine uterine secretions in dairy cows with endometritis using ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry. Res Vet Sci. 2021;139:51–8.

    Article  CAS  PubMed  Google Scholar 

  25. Valdes AM, Walter J, Segal E, Spector TD. Role of the gut microbiota in nutrition and health. BMJ. 2018;361:k2179.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Dupont G, Bordes SJ, Lachkar S, Wahl L, Iwanaga J, Loukas M, Tubbs RS. The effects of obesity on the human body part III: Cardiovascular, digestive, excretory, endocrine, and reproductive. Clin Anat. 2021;34(2):307–11.

    Article  PubMed  Google Scholar 

  27. Watanabe T, Minakata S, Kitagawa M. [Two cases of ureteral endometriosis]. Hinyokika Kiyo. 1989;35(2):315–21.

    CAS  PubMed  Google Scholar 

  28. Heuter KJ. Excretory urography. Clin Tech Small Anim Pract. 2005;20(1):39–45.

    Article  PubMed  Google Scholar 

  29. Ata B, Yildiz S, Turkgeldi E, Brocal VP, Dinleyici EC, Moya A, Urman B. The Endobiota Study: comparison of Vaginal, cervical and gut microbiota between women with stage 3/4 endometriosis and healthy controls. Sci Rep. 2019;9(1):2204.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Giannattasio-Ferraz S, Laguardia-Nascimento M, Gasparini MR, Leite LR, Araujo FMG, de Matos Salim AC, de Oliveira AP, Nicoli JR, de Oliveira GC, da Fonseca FG, et al. A common vaginal microbiota composition among breeds of Bos taurus indicus (Gyr and Nellore). Braz J Microbiol. 2019;50(4):1115–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Baker JM, Chase DM, Herbst-Kralovetz MM. Uterine microbiota: residents, tourists, or invaders? Front Immunol. 2018;9:208.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Sheldon IM, Cronin JG, Bromfield JJ. Tolerance and innate immunity shape the development of Postpartum Uterine Disease and the impact of endometritis in dairy cattle. Annu Rev Anim Biosci. 2019;7:361–84.

    Article  CAS  PubMed  Google Scholar 

  33. Jiang I, Yong PJ, Allaire C, Bedaiwy MA. Intricate connections between the Microbiota and Endometriosis. Int J Mol Sci 2021; 22(11).

  34. Wang ML, Liu MC, Xu J, An LG, Wang JF, Zhu YH. Uterine microbiota of dairy cows with clinical and subclinical endometritis. Front Microbiol. 2018;9:2691.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Svensson A, Brunkwall L, Roth B, Orho-Melander M, Ohlsson B. Associations between endometriosis and Gut Microbiota. Reprod Sci. 2021;28(8):2367–77.

    Article  CAS  PubMed  Google Scholar 

  36. McClure MW. The Vaginal Microbiome Related to Reproductive Traits in Beef Heifers. In: 2018;2018.

  37. Barinova VV, Kuznetsova NB, Bushtyreva IO, Dudurich VV. Uterine microbiome and immunohistochemical markers of chronic endometritis in recurrent pregnancy loss. Obstet Gynecol. 2022;4:84–94.

    Google Scholar 

  38. Cramer P, Bresalier RS. Gastrointestinal and hepatic complications of Immune Checkpoint inhibitors. Curr Gastroenterol Rep. 2017;19(1):3.

    Article  PubMed  Google Scholar 

  39. Zhang Q, Shen Q, Celestino J, Milam MR, Westin SN, Lacour RA, Meyer LA, Shipley GL, Davies PJ, Deng L, et al. Enhanced estrogen-induced proliferation in obese rat endometrium. Am J Obstet Gynecol. 2009;200(2):e186181–188.

    Article  Google Scholar 

  40. Jeon SJ, Lima FS, Vieira-Neto A, Machado VS, Lima SF, Bicalho RC, Santos JEP, Galvão KN. Shift of uterine microbiota associated with antibiotic treatment and cure of metritis in dairy cows. Vet Microbiol. 2018;214:132–9.

    Article  PubMed  Google Scholar 

  41. Karstrup CC, Klitgaard K, Jensen TK, Agerholm JS, Pedersen HG. Presence of bacteria in the endometrium and placentomes of pregnant cows. Theriogenology. 2017;99:41–7.

    Article  CAS  PubMed  Google Scholar 

  42. Moore SG, Ericsson AC, Poock SE, Melendez P, Lucy MC. Hot topic: 16S rRNA gene sequencing reveals the microbiome of the virgin and pregnant bovine uterus. J Dairy Sci. 2017;100(6):4953–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zelezniak A, Andrejev S, Ponomarova O, Mende DR, Bork P, Patil KR. Metabolic dependencies drive species co-occurrence in diverse microbial communities. Proceedings of the National Academy of Sciences 2015; 112(20):6449–6454.

  44. Zengler K, Zaramela LS. The social network of microorganisms - how auxotrophies shape complex communities. Nat Rev Microbiol. 2018;16(6):383–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Goshen T, Shpigel NY. Evaluation of intrauterine antibiotic treatment of clinical metritis and retained fetal membranes in dairy cows. Theriogenology. 2006;66(9):2210–8.

    Article  CAS  PubMed  Google Scholar 

  46. Liang H, Cai R, Li C, Glendon OHM, Chengcheng H, Yan H. High-throughput sequencing of 16S rRNA gene analysis reveals novel taxonomic diversity among vaginal microbiota in healthy and affected sows with endometritis. Res Vet Sci. 2022;143:33–40.

    Article  CAS  PubMed  Google Scholar 

  47. Magoč T, Salzberg SL. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinf (Oxford England). 2011;27(21):2957–63.

    Google Scholar 

  48. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30(15):2114–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, Fierer N, Peña AG, Goodrich JK, Gordon JI, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 2010;7(5):335–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat Methods. 2013;10(10):996–8.

    Article  CAS  PubMed  Google Scholar 

  51. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, Lesniewski RA, Oakley BB, Parks DH, Robinson CJ, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol. 2009;75(23):7537–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Douglas GM, Maffei VJ, Zaneveld JR, Yurgel SN, Brown JR, Taylor CM, Huttenhower C, Langille MGI. PICRUSt2 for prediction of metagenome functions. Nat Biotechnol. 2020;38(6):685–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Biagi E, Franceschi C, Rampelli S, Severgnini M, Ostan R, Turroni S, Consolandi C, Quercia S, Scurti M, Monti D, et al. Gut microbiota and Extreme Longevity. Curr Biol. 2016;26(11):1480–5.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Supported by Independent Innovation Project of Henan Academy of Agricultural Sciences (2018ZC56). The funding body played no role in the design of the study and collection, analysis, interpretation of data, and in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

ZHS conceived of the study, performed the research, and drafted the manuscript. YLL, ZHQ, XZY, YZW and BZ checked the heifers and collected the samples. XYM drafted the manuscript. FH revised the paper. WJW participated in the study’s design and assisted with the data analysis. TXD performed and analyzed the data, reviewed and edited the paper. All authors read and approved the final manuscript. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Wen-jia Wang or Ting-xian Deng.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the Animal Ethics Committee of Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences (Approval number SYXK 2020-0003), China. Experimental protocols for obtaining bovine clinical samples used in this study were carried out in strict accordance with the Animal Ethics Procedures and Guidelines of China.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Supplementary Material 2

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shi, Zh., Lan, Yl., Qiao, Zh. et al. Changes in fecal microbiota of dairy cows with and without endometritis. BMC Vet Res 21, 201 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12917-025-04580-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12917-025-04580-8

Keywords