1
|
Schmitz SS. Evidence-based use of biotics in the management of gastrointestinal disorders in dogs and cats. Vet Rec 2024; 195:26-32. [PMID: 39545596 DOI: 10.1002/vetr.4916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
The gut microbiome plays a vital role in the overall health of dogs and cats, particularly their gastrointestinal (GI) health. Imbalances in the gut microbiome, termed dysbiosis, are associated with a range of GI disorders, including diarrhoea and chronic enteropathy. Emerging evidence tentatively supports the therapeutic use of pre-, pro- and postbiotics (collectively called biotics) to restore gut homeostasis and manage these conditions, especially due to their potential antibiotic-sparing effects. This article explores their evidence-based use in the treatment of GI disorders in dogs and cats.
Collapse
|
2
|
Wilson SM, Swanson KS. The influence of 'biotics' on the gut microbiome of dogs and cats. Vet Rec 2024; 195:2-12. [PMID: 39545542 DOI: 10.1002/vetr.4914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
A global rise in pet ownership and an increasing tendency towards the humanisation of pets have resulted in a greater focus on improving animal health and longevity. These developments coincide with the increased recognition of the role of the gut microbiome in animal health. The gut microbiome has been shown to play a prominent role in gastrointestinal health, and it is becoming increasingly clear that these health benefits extend beyond the gut and into different physiological systems, such as the immune system. Dietary supplementation with products known as 'biotics', which include probiotics, prebiotics, synbiotics and postbiotics, is a strategy used to modify the gut microbiome and promote host health. Although biotics have been successfully used in companion animals, questions remain regarding appropriate biotic selection, mechanisms of action, optimum inclusion levels and safety. This review aims to summarise the effects of biotics on the gut microbiome of dogs and cats and assess their potential role in supporting gastrointestinal health.
Collapse
Affiliation(s)
- Sofia M Wilson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Kelly S Swanson
- Department of Animal Sciences, Department of Veterinary Medicine and Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
3
|
Schmid SM, Tolbert MK. Harnessing the microbiome: probiotics, antibiotics and their role in canine and feline gastrointestinal disease. Vet Rec 2024; 195:13-25. [PMID: 39545593 DOI: 10.1002/vetr.4915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Unfavourable alterations of the host microbial environment, known as dysbiosis, have been identified in many canine and feline gastrointestinal (GI) diseases. As a result, normalisation of microbial composition and function has become an important therapeutic target. Given the complex and individualistic interplay between the resident microbiota, host and environment, a multimodal approach is often necessary when addressing dysbiosis in dogs and cats with GI disease. Systemic antibiotics are often empirically used to treat acute and chronic GI diseases. However, with modern genomic techniques demonstrating the profound negative effect antibiotics can have on the GI microbiota and the rapid emergence of resistant bacteria globally, there has been an increased focus on identifying antibiotic alternatives for use in small animal practice. Biotics, such as prebiotics, probiotics and synbiotics, are of growing interest due to their potential supportive effect on the microbiota. This article reviews the evidence for the use of biotics in canine and feline GI disease, highlighting how judicious use of antibiotics and targeted probiotic supplementation can enhance patient outcomes by promoting a balanced gut microbial environment.
Collapse
Affiliation(s)
- Sarah M Schmid
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - M Katherine Tolbert
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
4
|
Rowe JC, Summers SC, Quimby JM, Winston JA. Fecal bile acid dysmetabolism and reduced ursodeoxycholic acid correlate with novel microbial signatures in feline chronic kidney disease. Front Microbiol 2024; 15:1458090. [PMID: 39498133 PMCID: PMC11532117 DOI: 10.3389/fmicb.2024.1458090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/16/2024] [Indexed: 11/07/2024] Open
Abstract
Background Microbial-derived secondary bile acids (SBAs) are reabsorbed and sensed via host receptors modulating cellular inflammation and fibrosis. Feline chronic kidney disease (CKD) occurs with progressive renal inflammation and fibrosis, mirroring the disease pathophysiology of human CKD patients. Methods Prospective cross-sectional study compared healthy cats (n = 6) with CKD (IRIS Stage 2 n = 17, Stage 3 or 4 n = 11). Single timepoint fecal samples from all cats underwent targeted bile acid metabolomics. 16S rRNA gene amplicon sequencing using DADA2 with SILVA taxonomy characterized the fecal microbiota. Results CKD cats had significantly reduced fecal concentrations (median 12.8 ng/mg, Mann-Whitney p = 0.0127) of the SBA ursodeoxycholic acid (UDCA) compared to healthy cats (median 39.4 ng/mg). Bile acid dysmetabolism characterized by <50% SBAs was present in 8/28 CKD and 0/6 healthy cats. Beta diversity significantly differed between cats with <50% SBAs and > 50% SBAs (PERMANOVA p < 0.0001). Twenty-six amplicon sequence variants (ASVs) with >97% nucleotide identity to Peptacetobacter hiranonis were identified. P. hiranonis combined relative abundance was significantly reduced (median 2.1%) in CKD cats with <50% SBAs compared to CKD cats with >50% SBAs (median 13.9%, adjusted p = 0.0002) and healthy cats with >50% SBAs (median 15.5%, adjusted p = 0.0112). P. hiranonis combined relative abundance was significantly positively correlated with the SBAs deoxycholic acid (Spearman r = 0.5218, adjusted p = 0.0407) and lithocholic acid (Spearman r = 0.5615, adjusted p = 0.0156). Three Oscillospirales ASVs and a Roseburia ASV were also identified as significantly correlated with fecal SBAs. Clinical and translational importance The gut-kidney axis mediated through microbial-derived SBAs appears relevant to the spontaneous animal CKD model of domestic cats. This includes reduced fecal concentrations of the microbial-derived SBA UDCA, known to regulate inflammation and fibrosis and be reno-protective. Microbes correlated with fecal SBAs include bai operon containing P. hiranonis, as well as members of Oscillospirales, which also harbor a functional bai operon. Ultimately, CKD cats represent a translational opportunity to study the role of SBAs in the gut-kidney axis, including the potential to identify novel microbial-directed therapeutics to mitigate CKD pathogenesis in veterinary patients and humans alike.
Collapse
Affiliation(s)
- John C. Rowe
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
| | - Stacie C. Summers
- Department of Clinical Sciences, Oregon State University Carlson College of Veterinary Medicine, Corvallis, OR, United States
| | - Jessica M. Quimby
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
| | - Jenessa A. Winston
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
| |
Collapse
|
5
|
Shah H, Trivedi M, Gurjar T, Sahoo DK, Jergens AE, Yadav VK, Patel A, Pandya P. Decoding the Gut Microbiome in Companion Animals: Impacts and Innovations. Microorganisms 2024; 12:1831. [PMID: 39338505 PMCID: PMC11433972 DOI: 10.3390/microorganisms12091831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
The changing notion of "companion animals" and their increasing global status as family members underscores the dynamic interaction between gut microbiota and host health. This review provides a comprehensive understanding of the intricate microbial ecology within companion animals required to maintain overall health and prevent disease. Exploration of specific diseases and syndromes linked to gut microbiome alterations (dysbiosis), such as inflammatory bowel disease, obesity, and neurological conditions like epilepsy, are highlighted. In addition, this review provides an analysis of the various factors that impact the abundance of the gut microbiome like age, breed, habitual diet, and microbe-targeted interventions, such as probiotics. Detection methods including PCR-based algorithms, fluorescence in situ hybridisation, and 16S rRNA gene sequencing are reviewed, along with their limitations and the need for future advancements. Prospects for longitudinal investigations, functional dynamics exploration, and accurate identification of microbial signatures associated with specific health problems offer promising directions for future research. In summary, it is an attempt to provide a deeper insight into the orchestration of multiple microbial species shaping the health of companion animals and possible species-specific differences.
Collapse
Affiliation(s)
- Harsh Shah
- TREE Lab, Department of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara 391410, India (M.T.)
| | - Mithil Trivedi
- TREE Lab, Department of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara 391410, India (M.T.)
| | - Tejas Gurjar
- TREE Lab, Department of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara 391410, India (M.T.)
| | - Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
| | - Albert E. Jergens
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
| | - Virendra Kumar Yadav
- Marwadi University Research Center, Department of Microbiology, Faculty of Sciences, Marwadi University, Rajkot 360003, India;
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan 384265, India;
| | - Parth Pandya
- TREE Lab, Department of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara 391410, India (M.T.)
| |
Collapse
|
6
|
Wang W, Dong H, Chang X, Chen Q, Wang L, Chen S, Chen L, Wang R, Ge S, Wang P, Li Y, Liu S, Xiong W. Bifidobacterium lactis and Lactobacillus plantarum Enhance Immune Function and Antioxidant Capacity in Cats through Modulation of the Gut Microbiota. Antioxidants (Basel) 2024; 13:764. [PMID: 39061833 PMCID: PMC11273429 DOI: 10.3390/antiox13070764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/15/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Gastrointestinal (GI) afflictions are prevalent among the feline population, wherein the intricacies of the gut microbiome exert a profound influence on their overall health. Alterations within this microbial consortium can precipitate a cascade of physiological changes, notably in immune function and antioxidant capacity. This research investigated the impact of Bifidobacterium lactis (B. lactis) and Lactobacillus plantarum (L. plantarum) on cats' GI health, exploring the effects of probiotic supplementation on the intestinal ecosystem using 16S rRNA gene sequencing. The findings demonstrated a significant improvement in gut barrier function by reducing plasma concentrations of D-lactate (D-LA) by 30.38% and diamine oxidase (DAO) by 22.68%, while increasing the population of beneficial bacteria such as Lactobacillus. There was a notable 25% increase in immunoglobulin A (IgA) levels, evidenced by increases of 19.13% in catalase (CAT), 23.94% in superoxide dismutase (SOD), and 21.81% in glutathione peroxidase (GSH-Px). Further analysis revealed positive correlations between Lactobacillus abundance and IgA, CAT, and total antioxidant capacity (T-AOC) levels. These correlations indicate that B. lactis and L. plantarum enhance feline immune and antioxidant functions by increasing the abundance of beneficial Lactobacillus in the GI tract. These findings provide a foundation for probiotic interventions aimed at enhancing health and disease resistance in feline populations.
Collapse
Affiliation(s)
- Weiwei Wang
- Food Laboratory of Zhongyuan, Luohe 462300, China; (W.W.); (H.D.); (X.C.); (Q.C.); (S.C.); (L.C.)
| | - Hao Dong
- Food Laboratory of Zhongyuan, Luohe 462300, China; (W.W.); (H.D.); (X.C.); (Q.C.); (S.C.); (L.C.)
| | - Xiaohan Chang
- Food Laboratory of Zhongyuan, Luohe 462300, China; (W.W.); (H.D.); (X.C.); (Q.C.); (S.C.); (L.C.)
| | - Qianqian Chen
- Food Laboratory of Zhongyuan, Luohe 462300, China; (W.W.); (H.D.); (X.C.); (Q.C.); (S.C.); (L.C.)
| | - Longjiao Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (L.W.); (R.W.); (S.G.); (P.W.); (Y.L.); (S.L.)
| | - Shuxing Chen
- Food Laboratory of Zhongyuan, Luohe 462300, China; (W.W.); (H.D.); (X.C.); (Q.C.); (S.C.); (L.C.)
| | - Lishui Chen
- Food Laboratory of Zhongyuan, Luohe 462300, China; (W.W.); (H.D.); (X.C.); (Q.C.); (S.C.); (L.C.)
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (L.W.); (R.W.); (S.G.); (P.W.); (Y.L.); (S.L.)
| | - Shaoyang Ge
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (L.W.); (R.W.); (S.G.); (P.W.); (Y.L.); (S.L.)
| | - Pengjie Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (L.W.); (R.W.); (S.G.); (P.W.); (Y.L.); (S.L.)
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (L.W.); (R.W.); (S.G.); (P.W.); (Y.L.); (S.L.)
| | - Siyuan Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (L.W.); (R.W.); (S.G.); (P.W.); (Y.L.); (S.L.)
| | - Wei Xiong
- Food Laboratory of Zhongyuan, Luohe 462300, China; (W.W.); (H.D.); (X.C.); (Q.C.); (S.C.); (L.C.)
| |
Collapse
|
7
|
Drut A, Mkaouar H, Kriaa A, Mariaule V, Akermi N, Méric T, Sénécat O, Maguin E, Hernandez J, Rhimi M. Gut microbiota in cats with inflammatory bowel disease and low-grade intestinal T-cell lymphoma. Front Microbiol 2024; 15:1346639. [PMID: 38812688 PMCID: PMC11133722 DOI: 10.3389/fmicb.2024.1346639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
In cats and humans, several physiological and environmental factors have been shown to alter the gut microbiota of healthy individuals. Cats share several diseases with humans such as inflammatory bowel diseases and low-grade intestinal T-cell lymphoma. The physiopathology of these chronic enteropathies is poorly understood but may involve disequilibrium of the gut microbiota composition and disruption of normal microbiome activity profiles. These disorders are increasingly diagnosed in the feline species due to improved medicalization and easier access to endoscopy in veterinary practice. This review addresses the current data on the gut microbiota of cats in health and in chronic enteropathies. Such functional analysis will help the advancement of innovative diagnostic tools and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Amandine Drut
- MIHA Team, INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Oniris VetAgroBio Nantes, Université de Nantes, Nantes, France
| | - Héla Mkaouar
- MIHA Team, INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Aicha Kriaa
- MIHA Team, INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Vincent Mariaule
- MIHA Team, INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Nizar Akermi
- MIHA Team, INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Tristan Méric
- MIHA Team, INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Oniris VetAgroBio Nantes, Université de Nantes, Nantes, France
| | - Odile Sénécat
- Oniris VetAgroBio Nantes, Université de Nantes, Nantes, France
| | - Emmanuelle Maguin
- MIHA Team, INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Juan Hernandez
- MIHA Team, INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Oniris VetAgroBio Nantes, Université de Nantes, Nantes, France
| | - Moez Rhimi
- MIHA Team, INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
8
|
Rowe JC, Winston JA. Collaborative Metabolism: Gut Microbes Play a Key Role in Canine and Feline Bile Acid Metabolism. Vet Sci 2024; 11:94. [PMID: 38393112 PMCID: PMC10892723 DOI: 10.3390/vetsci11020094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Bile acids, produced by the liver and secreted into the gastrointestinal tract, are dynamic molecules capable of impacting the overall health of dogs and cats in many contexts. Importantly, the gut microbiota metabolizes host primary bile acids into chemically distinct secondary bile acids. This review explores the emergence of new literature connecting microbial-derived bile acid metabolism to canine and feline health and disease. Moreover, this review highlights multi-omic methodologies for translational research as an area for continued growth in veterinary medicine aimed at accelerating microbiome science and medicine as it pertains to bile acid metabolism in dogs and cats.
Collapse
Affiliation(s)
- John C. Rowe
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH 43210, USA;
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH 43210, USA
| | - Jenessa A. Winston
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH 43210, USA;
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH 43210, USA
| |
Collapse
|
9
|
Langon X. Validation of method for faecal sampling in cats and dogs for faecal microbiome analysis. BMC Vet Res 2023; 19:274. [PMID: 38102642 PMCID: PMC10724939 DOI: 10.1186/s12917-023-03842-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/03/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Reproducible and reliable studies of cat and dog faecal microbiomes are dependent on many methodology-based variables including how the faecal stools are sampled and stored prior to processing. The current study aimed to establish an appropriate method for sampling and storing faecal stools from cats and dogs which may also be applied to privately-owned pets. The approach investigated the effects of storing faeces for up to 12 h at room temperature and sampling from various locations within the stool in terms of microbial diversity, relative taxa abundances and DNA yield. Faeces were collected from 10 healthy cats and 10 healthy dogs and stored at room temperature (20 °C). Samples were taken from various locations within the stool (the first emitted part (i), the middle (ii) and the last emitted end (iii), at either surface or core) at 0, 0.5, 1, 2, 3, 6 and 12 h, stabilised and stored at -80 °C. DNA was extracted from all samples, using Illumina NovaSeq. RESULTS Faecal bacterial composition of dogs and cats shown no statistically significant differences in alpha diversity. Bacteroidetes, Firmicutes, Proteobacteria and Actinobacteria were the most prevalent phyla. Cat and dog samples were characterized by a dominance of Prevotella, and a lack of Fusobacterium in feline stools. Room temperature storage of cat and dog faecal samples generally had no significant effect on alpha diversity, relative taxa abundance or DNA yield for up to 12 h. Sampling from regions i, ii or iii of the stool at the surface or core did not significantly influence the outcome. However, surface cat faecal samples stored at room temperature for 12 h showed a significant increase in two measures of alpha diversity and there was a tendency for a similar effect in dogs. When comparing samples with beta diversity measures, it appeared that for dog and cat samples, individual effect has the strongest impact on the observed microbial diversity (R2 0.64 and 0.88), whereas sampling time, depth and horizontal locations significantly affected the microbial diversity but with less impact. CONCLUSION Cat and dog faeces were stable at room temperature for up to 12 h, with no significant changes in alpha diversity, relative taxa abundance and DNA concentration. Beta diversity analysis demonstrated that despite an impact of the sampling storing time and the surface of the sampling, we preserved the identity of the microbial structure linked to the individual. Finally, the data suggest that faecal stools stored for > 6 h at room temperature should be sampled at the core, not the surface.
Collapse
Affiliation(s)
- Xavier Langon
- Royal Canin Sas, 650 avenue de la Petite Camargue, AIMARGUES Cedex, CS, 10309, 30470, France.
| |
Collapse
|
10
|
Yang Q, Wu Z. Gut Probiotics and Health of Dogs and Cats: Benefits, Applications, and Underlying Mechanisms. Microorganisms 2023; 11:2452. [PMID: 37894110 PMCID: PMC10609632 DOI: 10.3390/microorganisms11102452] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Pets (mostly domestic dogs and cats) play an important role in the daily lives of humans and their health has attracted growing attention from pet owners. The intestinal microbiota, a complex microbial community with barrier-protective, nutritional, metabolic, and immunological functions, is integral to host health. Dysbiosis has been related to a variety of diseases in humans and animals. Probiotics have been used in functional foods and dietary supplements to modulate intestinal microbiota and promote host health, which has been introduced in pet dogs and cats in recent years. Various canine- and feline-derived probiotic strains have been isolated and characterized. The administration of probiotics has shown positive effects on the gut health and can alleviate some intestinal diseases and disorders in dogs and cats, although the underlying mechanisms are largely unresolved. In this review, we summarize the current knowledge on the benefits of probiotics and discuss their possible mechanisms in dogs and cats in order to provide new insights for the further development and application of probiotics in pets.
Collapse
Affiliation(s)
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, China Agricultural University, Beijing 100193, China;
| |
Collapse
|
11
|
Rojas CA, Entrolezo Z, Jarett JK, Jospin G, Kingsbury DD, Martin A, Eisen JA, Ganz HH. Microbiome Responses to Fecal Microbiota Transplantation in Cats with Chronic Digestive Issues. Vet Sci 2023; 10:561. [PMID: 37756083 PMCID: PMC10537086 DOI: 10.3390/vetsci10090561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/25/2023] [Accepted: 09/02/2023] [Indexed: 09/28/2023] Open
Abstract
There is growing interest in the application of fecal microbiota transplants (FMTs) in small animal medicine, but there are few published studies that have tested their effects in the domestic cat (Felis catus). Here we use 16S rRNA gene sequencing to examine fecal microbiome changes in 46 domestic cats with chronic digestive issues that received FMTs using lyophilized stool that was delivered in oral capsules. Fecal samples were collected from FMT recipients before and two weeks after the end of the full course of 50 capsules, as well as from their stool donors (N = 10), and other healthy cats (N = 113). The fecal microbiomes of FMT recipients varied with host clinical signs and dry kibble consumption, and shifts in the relative abundances of Clostridium, Collinsella, Megamonas, Desulfovibrio and Escherichia were observed after FMT. Overall, donors shared 13% of their bacterial amplicon sequence variants (ASVs) with FMT recipients and the most commonly shared ASVs were classified as Prevotella 9, Peptoclostridium, Bacteroides, and Collinsella. Lastly, the fecal microbiomes of cats with diarrhea became more similar to the microbiomes of age-matched and diet-matched healthy cats compared to cats with constipation. Overall, our results suggest that microbiome responses to FMT may be modulated by the FMT recipient's initial presenting clinical signs, diet, and their donor's microbiome.
Collapse
Affiliation(s)
- Connie A. Rojas
- Genome Center, University of California, Davis, CA 95616, USA; (C.A.R.); (J.A.E.)
- Department of Evolution and Ecology, University of California, Davis, CA 95616, USA
| | - Zhandra Entrolezo
- AnimalBiome, Oakland, CA 94609, USA; (Z.E.); (J.K.J.); (G.J.); (A.M.)
| | - Jessica K. Jarett
- AnimalBiome, Oakland, CA 94609, USA; (Z.E.); (J.K.J.); (G.J.); (A.M.)
| | - Guillaume Jospin
- AnimalBiome, Oakland, CA 94609, USA; (Z.E.); (J.K.J.); (G.J.); (A.M.)
| | - Dawn D. Kingsbury
- AnimalBiome, Oakland, CA 94609, USA; (Z.E.); (J.K.J.); (G.J.); (A.M.)
| | - Alex Martin
- AnimalBiome, Oakland, CA 94609, USA; (Z.E.); (J.K.J.); (G.J.); (A.M.)
| | - Jonathan A. Eisen
- Genome Center, University of California, Davis, CA 95616, USA; (C.A.R.); (J.A.E.)
- Department of Evolution and Ecology, University of California, Davis, CA 95616, USA
| | - Holly H. Ganz
- AnimalBiome, Oakland, CA 94609, USA; (Z.E.); (J.K.J.); (G.J.); (A.M.)
| |
Collapse
|
12
|
Stavroulaki EM, Suchodolski JS, Xenoulis PG. Effects of antimicrobials on the gastrointestinal microbiota of dogs and cats. Vet J 2023; 291:105929. [PMID: 36427604 DOI: 10.1016/j.tvjl.2022.105929] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 11/12/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Among several environmental factors, exposure to antimicrobials has been in the spotlight as a cause of profound and long-term disturbance of the intestinal microbiota. Antimicrobial-induced dysbiosis is a general term and includes decreases in microbial richness and diversity, loss of beneficial bacterial groups, blooms of intestinal pathogens and alterations in the metabolic functions and end-products of the microbiota. Mounting evidence from human and experimental animal studies suggest an association between antimicrobial-induced dysbiosis and susceptibility to gastrointestinal, metabolic, endocrine, immune and neuropsychiatric diseases. These associations are commonly stronger after early life exposure to antimicrobials, a period during which maturation of the microbiota and immune system take place in parallel. In addition, these associations commonly become stronger as the number of antimicrobial courses increases. The repeatability of these findings among different studies as well as the presence of a dose-dependent relationship between antimicrobial exposure and disease development collectively require careful consideration of the need for antimicrobial use. There are limited studies are available in dogs and cats regarding the long-term effects of antimicrobials on the microbiota and subsequent susceptibility to diseases. This review discusses the effects of antimicrobials on the gastrointestinal microbiota and the most important associations between antimicrobial-induced dysbiosis and diseases in humans, dogs, and cats.
Collapse
Affiliation(s)
- Evangelia M Stavroulaki
- Clinic of Medicine, Faculty of Veterinary Science, University of Thessaly, Karditsa 43131, Greece.
| | - Jan S Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station 77845, TX, USA
| | - Panagiotis G Xenoulis
- Clinic of Medicine, Faculty of Veterinary Science, University of Thessaly, Karditsa 43131, Greece; Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station 77845, TX, USA
| |
Collapse
|
13
|
Vojtkovská V, Kaluža M, Voslářová E, Večerek V, Tomečková L. The morbidity and the spectrum of applied drugs in shelter cats in the Czech Republic. Front Vet Sci 2022; 9:1025197. [PMID: 36544553 PMCID: PMC9760937 DOI: 10.3389/fvets.2022.1025197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
Maintaining appropriate living conditions and good health of the animals should be one of the main priorities of cat shelters. The aim of this study was to assess the morbidity of shelter cats on the basis of analysis of the shelter health records in terms of the nature and quantity of medicaments and other supportive products administered in two no-kill shelters in the Czech Republic. The subject of the study were the health records of 1,884 cats, which were admitted to the selected shelters from 1.1.2013 to 31.12.2021 and their stay in the shelter was terminated in the monitored period. More than half of all cats whose records were analyzed had at least one health record made during their stay in the shelter. The overall length of stay of cats in the shelter (regardless of the outcome) in which one or more health records were found was significantly longer compared to the length of stay of cats without any health records. The highest number of health records was found in kittens <6 months old. Antibiotics, antiparasitics, and complementary/protective substances were the most used among all administered drugs. Broad-spectrum antibiotics have been administered to cats most often. In terms of classification of antibiotics according to the active substance, the most used antibiotics were penicillins, tetracyclines, and fluoroquinolones. Antibiotics were mostly used to treat diseases related to the upper and lower respiratory tract and their symptoms and gastrointestinal problems. The findings of the study contribute to understanding health problems and approaches to treating the shelter cats.
Collapse
Affiliation(s)
- Veronika Vojtkovská
- Department of Animal Protection and Welfare and Veterinary Public Health, University of Veterinary Sciences Brno, Brno, Czechia,*Correspondence: Veronika Vojtkovská
| | - Michal Kaluža
- Department of Animal Protection and Welfare and Veterinary Public Health, University of Veterinary Sciences Brno, Brno, Czechia
| | - Eva Voslářová
- Department of Animal Protection and Welfare and Veterinary Public Health, University of Veterinary Sciences Brno, Brno, Czechia
| | - Vladimír Večerek
- Department of Animal Protection and Welfare and Veterinary Public Health, University of Veterinary Sciences Brno, Brno, Czechia
| | - Lenka Tomečková
- Department of Public Veterinary Medicine and Animal Welfare, The University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| |
Collapse
|
14
|
Gookin JL, Strong SJ, Bruno-Bárcena JM, Stauffer SH, Williams S, Wassack E, Azcarate-Peril MA, Estrada M, Seguin A, Balzer J, Davidson G. Randomized placebo-controlled trial of feline-origin Enterococcus hirae probiotic effects on preventative health and fecal microbiota composition of fostered shelter kittens. Front Vet Sci 2022; 9:923792. [PMID: 36467638 PMCID: PMC9714445 DOI: 10.3389/fvets.2022.923792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 11/02/2022] [Indexed: 10/29/2023] Open
Abstract
Introduction Diarrhea is the second most common cause of mortality in shelter kittens. Studies examining prevention strategies in this population are lacking. Probiotics are of particular interest but studies in cats are largely limited to healthy adults or those with induced disease. Only one study in domestic cats describes the use of host-derived bacteria as a probiotic. We previously identified Enterococcus hirae as a dominant species colonizing the small intestinal mucosa in healthy shelter kittens. Oral administration of a probiotic formulation of kitten-origin E. hirae (strain 1002-2) mitigated the increase in intestinal permeability and fecal water loss resulting from experimental enteropathogenic E. coli infection in purpose-bred kittens. Based on these findings, we hypothesized that administration of kitten-origin E. hirae to weaned fostered shelter kittens could provide a measurable preventative health benefit. Methods We conducted a randomized, placebo-controlled, blinded clinical trial to determine the impact of a freeze-dried E. hirae probiotic on body weight gain, incidence of diarrhea, carriage of potential diarrheal pathogens, and composition of the intestinal microbiota in weaned fostered shelter kittens. Results One-hundred thirty kittens completed the study. Fifty-eight kittens received the probiotic and 72 received the placebo. There were no significant differences in age, weight upon initiation of the study, number of days in the study, average daily gain in body weight, or weight at completion of the study. Kittens treated with E. hirae were 3.4 times less likely to develop diarrhea compared to kittens treated with placebo (odds ratio = 0.294, 95% CI 0.109-0.792, p = 0.022). A significant impact of E. hirae was not observed on the presence or abundance of 30 different bacterial, viral, protozoal, fungal, algal, and parasitic agents in feces examined by qPCR. With exception to a decrease in Megamonas, administration of the E. hirae probiotic did not alter the predominant bacterial phyla present in feces based on 16S rRNA gene amplicon sequencing. Discussion Decreased incidence of diarrhea associated with preventative administration of E. hirae to foster kittens supports a rationale for use of E. hirae for disease prevention in this young population at high risk for intestinal disease though additional studies are warranted.
Collapse
Affiliation(s)
- Jody L. Gookin
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Sandra J. Strong
- Department of Environmental Services, Wake County Animal Center, Raleigh, NC, United States
- Orange County Animal Services, Chapel Hill, NC, United States
| | - José M. Bruno-Bárcena
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, United States
| | - Stephen H. Stauffer
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Shelby Williams
- Veterinary Hospital Pharmacy, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- University of Wisconsin Veterinary Care, Madison, WI, United States
| | - Erica Wassack
- Veterinary Hospital Pharmacy, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS, United States
| | - M. Andrea Azcarate-Peril
- Division of Gastroenterology and Hepatology, and UNC Microbiome Core, Department of Medicine, Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Marko Estrada
- IDEXX Laboratories, Inc., West Sacramento, CA, United States
| | - Alexis Seguin
- IDEXX Laboratories, Inc., West Sacramento, CA, United States
| | - Joerg Balzer
- Vet Med Labor GmbH Division, IDEXX Laboratories, Inc., Kornwestheim, Germany
| | - Gigi Davidson
- Veterinary Hospital Pharmacy, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
15
|
Whole-Genome Shotgun Metagenomic Sequencing Reveals Distinct Gut Microbiome Signatures of Obese Cats. Microbiol Spectr 2022; 10:e0083722. [PMID: 35467389 PMCID: PMC9241680 DOI: 10.1128/spectrum.00837-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Overweight and obesity are growing health problems in domestic cats, increasing the risks of insulin resistance, lipid dyscrasias, neoplasia, cardiovascular disease, and decreasing longevity. The signature of obesity in the feline gut microbiota has not been studied at the whole-genome metagenomic level. We performed whole-genome shotgun metagenomic sequencing in the fecal samples of eight overweight/obese and eight normal cats housed in the same research environment. We obtained 271 Gbp of sequences and generated a 961-Mbp de novo reference contig assembly, with 1.14 million annotated microbial genes. In the obese cat microbiome, we discovered a significant reduction in microbial diversity (P < 0.01) and Firmicutes abundance (P = 0.005), as well as decreased Firmicutes/Bacteroidetes ratios (P = 0.02), which is the inverse of obese human/mouse microbiota. Linear discriminant analysis and quantitative PCR (qPCR) validation revealed significant increases of Bifidobacterium sp., Olsenella provencensis, Dialister sp.CAG:486, and Campylobacter upsaliensis as the hallmark of obese microbiota among 400 enriched species, whereas 1,525 bacterial species have decreased abundance in the obese microbiome. Phascolarctobacterium succinatutens and an uncharacterized Erysipelotrichaceae bacterium are highly abundant (>0.05%) in the normal gut with over 400-fold depletion in the obese microbiome. Fatty acid synthesis-related pathways are significantly overrepresented in the obese compared with the normal cat microbiome. In conclusion, we discovered dramatically decreased microbial diversity in obese cat gut microbiota, suggesting potential dysbiosis. A panel of seven significantly altered, highly abundant species can serve as a microbiome indicator of obesity. Our findings in the obese cat microbiome composition, abundance, and functional capacities provide new insights into feline obesity. IMPORTANCE Obesity affects around 45% of domestic cats, and licensed drugs for treating feline obesity are lacking. Physical exercise and calorie restrictions are commonly used for weight loss but with limited efficacy. Through comprehensive analyses of normal and obese cat gut bacteria flora, we identified dramatic shifts in the obese gut microbiome, including four bacterial species significantly enriched and two species depleted in the obese cats. The key bacterial community and functional capacity alterations discovered from this study will inform new weight management strategies for obese cats, such as evaluations of specific diet formulas that alter the microbiome composition, and the development of prebiotics and probiotics that promote the increase of beneficial species and the depletion of obesity-associated species. Interestingly, these bacteria identified in our study were also reported to affect the weight loss success in human patients, suggesting translational potential in human obesity.
Collapse
|
16
|
Tong G, Qian H, Li D, Li J, Chen J, Li X. Establishment and evaluation of a specific antibiotic-induced inflammatory bowel disease model in rats. PLoS One 2022; 17:e0264194. [PMID: 35192646 PMCID: PMC8863245 DOI: 10.1371/journal.pone.0264194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 02/06/2022] [Indexed: 12/03/2022] Open
Abstract
Physical and chemical methods for generating rat models of enteritis have been established; however, antibiotic induction has rarely been used for this purpose. The present study aimed to establish and evaluate a rat model of inflammatory bowel disease (IBD) using antibiotics. A total of 84 Sprague-Dawley (SD) rats were divided into the following groups, according to the dosage and method of administration of the antibiotics: A, control; B, low-dose clindamycin; C, medium-dose clindamycin; D, high-dose clindamycin; E, low-dose clindamycin, ampicillin and streptomycin; F, medium-dose clindamycin, ampicillin and streptomycin; and G, high-dose clindamycin, ampicillin and streptomycin. Antibiotic administration was stopped on day 7; the modeling period covered days 1-7, and the recovery period covered days 8-15. Half of the animals were dissected on day 11, with the remaining animals dissected on day 15. Food and water intake, body weight and fecal weight were recorded. Intestinal flora was analyzed via microbial culture and quantitative PCR. The content of TNF-α, IL1-β, IL-6 and C-reactive protein (CRP) was assessed in abdominal aorta blood. Colonic and rectal tissues were examined pathologically via hematoxylin-eosin staining to assess leukocyte infiltration and intestinal mucosal changes as indicators of inflammation. Rat weight, food intake, water intake and 2-h fecal weight were significantly different across the experimental groups (P = 0.040, P = 0.016, P<0.001 and P = 0.009, respectively). Microbial cultures revealed no significant differences between group A and B,C (P = 0.546,0.872) but significant differences betwenn group A and the other experimental groups (all P<0.001). Furthermore, significant differences in the levels of Bacteroides, Faecalibacterium prausnitzii and Dialister invisus on day 4 between groups A, C and F (P = 0.033, P = 0.025 and P = 0.034, respectively). Significant differences were detected in the levels of TNF-α, IL1-β, IL-6 and CRP between the groups (all P<0.001). The colonic and rectal pathological inflammation scores of the experimental groups were significantly different compared with group A (B vs. A, P = 0.002; others, all P<0.001). These findings indicated that an antibiotic-induced IBD model was successfully established in SD rats; this animal model may serve as a useful model for clinical IBD research.
Collapse
Affiliation(s)
- Guojun Tong
- Departments of General Surgery, Huzhou Central Hospital, Huzhou, Zhejiang, China
- Central Laboratory, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Hai Qian
- Departments of General Surgery, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Dongli Li
- Central Laboratory, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Jing Li
- Central Laboratory, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Jing Chen
- Central Laboratory, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Xiongfeng Li
- Orthopedic Surgery, Huzhou Central Hospital, Huzhou, Zhejiang, China
| |
Collapse
|
17
|
Rutherford S, Gaschen F, Husnik R, Fletcher J, Gaschen L. Ultrasonographic evaluation of the effects of azithromycin on antral motility and gastric emptying in healthy cats. J Vet Intern Med 2022; 36:508-514. [PMID: 35150012 PMCID: PMC8965205 DOI: 10.1111/jvim.16385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 11/29/2022] Open
Abstract
Background Erythromycin, a macrolide antibiotic with motilin agonist properties, shortens gastric emptying (GE) time in healthy cats. Azithromycin, another macrolide antibiotic, is effective for treatment of gastric paresis in people. Objectives To evaluate the effects of azithromycin on GE and gastric motility in healthy cats in comparison with erythromycin (positive control) and placebo. Animals Eight healthy purpose‐bred cats. Methods Prospective, blinded, crossover study. Cats received either azithromycin (3.5 mg/kg PO q24h), erythromycin (1 mg/kg PO q8h), or placebo for 24 hours before and during evaluation of GE. A validated method using ultrasound for sequential measurements of antral area as well as amplitude and frequency of contractions was used to assess GE and evaluate gastric antral motility postprandially over an 8‐hour period. Results GE was significantly faster (P < .05) after administration of azithromycin and erythromycin when compared to placebo in the late phase of fractional emptying from 75% (mean ± SD: 327 ± 51 minutes, 327 ± 22 minutes, and 367 ± 29 minutes, respectively), to 95% fractional emptying (399 ± 52 minutes, 404 ± 11 minutes, and 444 ± 24 minutes, respectively). The drugs had no significant effect on antral motility variables at any time point. Conclusions and Clinical Importance Azithromycin and erythromycin shorten GE time in a comparable manner in healthy cats. Evaluation of their efficacy in cats with gastric dysmotility is warranted.
Collapse
Affiliation(s)
- Stephanie Rutherford
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Frederic Gaschen
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Roman Husnik
- Department of Veterinary Clinical Sciences, Purdue University College of Veterinary Medicine, West Lafayette, Indiana, USA
| | | | - Lorrie Gaschen
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
18
|
The Serum and Fecal Metabolomic Profiles of Growing Kittens Treated with Amoxicillin/Clavulanic Acid or Doxycycline. Animals (Basel) 2022; 12:ani12030330. [PMID: 35158655 PMCID: PMC8833518 DOI: 10.3390/ani12030330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary This study investigated the impact of antibiotic treatment οn the serum and fecal metabolome (the collection of all small molecules produced by the gut bacteria and the host) of young cats. Thirty 2-month-old cats with an upper respiratory tract infection were treated with either amoxicillin/clavulanic acid for 20 days or doxycycline for 28 days. In addition, another 15 control cats that did not receive antibiotics were included. Blood was collected on days 0 (before treatment), 20/28 (last day of treatment), and 300 (10 months after the end of treatment), while feces were collected on days 0, 20/28, 60, 120, and 300. Seven serum and fecal metabolites differed between cats treated with antibiotics and control cats at the end of treatment period. Ten months after treatment, no metabolites differed from healthy cats, suggesting that amoxicillin/clavulanic acid or doxycycline treatment only temporarily affects the abundance of the serum and fecal metabolome. Abstract The long-term impact of antibiotics on the serum and fecal metabolome of kittens has not yet been investigated. Therefore, the objective of this study was to evaluate the serum and fecal metabolome of kittens with an upper respiratory tract infection (URTI) before, during, and after antibiotic treatment and compare it with that of healthy control cats. Thirty 2-month-old cats with a URTI were randomly assigned to receive either amoxicillin/clavulanic acid for 20 days or doxycycline for 28 days, and 15 cats of similar age were enrolled as controls. Fecal samples were collected on days 0, 20/28, 60, 120, and 300, while serum was collected on days 0, 20/28, and 300. Untargeted and targeted metabolomic analyses were performed on both serum and fecal samples. Seven metabolites differed significantly in antibiotic-treated cats compared to controls on day 20/28, with two differing on day 60, and two on day 120. Alterations in the pattern of serum amino acids, antioxidants, purines, and pyrimidines, as well as fecal bile acids, sterols, and fatty acids, were observed in antibiotic-treated groups that were not observed in control cats. However, the alterations caused by either amoxicillin/clavulanic acid or doxycycline of the fecal and serum metabolome were only temporary and were resolved by 10 months after their withdrawal.
Collapse
|
19
|
Short- and long-term effects of amoxicillin/clavulanic acid or doxycycline on the gastrointestinal microbiome of growing cats. PLoS One 2021; 16:e0253031. [PMID: 34910719 PMCID: PMC8673677 DOI: 10.1371/journal.pone.0253031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 10/18/2021] [Indexed: 02/07/2023] Open
Abstract
Antibiotic treatment in early life influences gastrointestinal (GI) microbial composition and function. In humans, the resultant intestinal dysbiosis is associated with an increased risk for certain diseases later in life. The objective of this study was to determine the temporal effects of antibiotic treatment on the GI microbiome of young cats. Fecal samples were collected from cats randomly allocated to receive either amoxicillin/clavulanic acid (20 mg/kg q12h) for 20 days (AMC group; 15 cats) or doxycycline (10 mg/kg q24h) for 28 days (DOX group;15 cats) as part of the standard treatment of upper respiratory tract infection. In addition, feces were collected from healthy control cats (CON group;15 cats). All cats were approximately two months of age at enrolment. Samples were collected on days 0 (baseline), 20 or 28 (AMC and DOX, respectively; last day of treatment), 60, 120, and 300. DNA was extracted and sequencing of the 16S rRNA gene and qPCR assays were performed. Fecal microbial composition was different on the last day of treatment for AMC cats, and 1 month after the end of antibiotic treatment for DOX cats, compared to CON cats. Species richness was significantly greater in DOX cats compared to CON cats on the last day of treatment. Abundance of Enterobacteriales was increased, and that of Erysipelotrichi was decreased in cats of the AMC group on the last day of treatment compared to CON cats. The abundance of the phylum Proteobacteria was increased in cats of the DOX group on days 60 and 120 compared to cats of the CON group. Only minor differences in abundances between the treatment groups and the control group were present on day 300. Both antibiotics appear to delay the developmental progression of the microbiome, and this effect is more profound during treatment with amoxicillin/clavulanic acid and one month after treatment with doxycycline. Future studies are required to determine if these changes influence microbiome function and whether they have possible effects on disease susceptibility in cats.
Collapse
|
20
|
Impact of Antibiotic Therapies on Resistance Genes Dynamic and Composition of the Animal Gut Microbiota. Animals (Basel) 2021; 11:ani11113280. [PMID: 34828011 PMCID: PMC8614244 DOI: 10.3390/ani11113280] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/03/2021] [Accepted: 11/11/2021] [Indexed: 11/16/2022] Open
Abstract
Antibiotics are major disruptors of the gastrointestinal microbiota, depleting bacterial species beneficial for the host health and favoring the emergence of potential pathogens. Furthermore, the intestine is a reactor of antibiotic resistance emergence, and the presence of antibiotics exacerbates the selection of resistant bacteria that can disseminate in the environment and propagate to further hosts. We reviewed studies analyzing the effect of antibiotics on the intestinal microbiota and antibiotic resistance conducted on animals, focusing on the main food-producing and companion animals. Irrespective of antibiotic classes and animal hosts, therapeutic dosage decreased species diversity and richness favoring the bloom of potential enteropathogens and the selection of antibiotic resistance. These negative effects of antibiotic therapies seem ineluctable but often were mitigated when an antibiotic was administered by parenteral route. Sub-therapeutic dosages caused the augmentation of taxa involved in sugar metabolism, suggesting a link with weight gain. This result should not be interpreted positively, considering that parallel information on antibiotic resistance selection was rarely reported and selection of antibiotic resistance is known to occur also at low antibiotic concentration. However, studies on the effect of antibiotics as growth promoters put the basis for understanding the gut microbiota composition and function in this situation. This knowledge could inspire alternative strategies to antibiotics, such as probiotics, for improving animal performance. This review encompasses the analysis of the main animal hosts and all antibiotic classes, and highlights the future challenges and gaps of knowledge that should be filled. Further studies are necessary for elucidating pharmacodynamics in animals in order to improve therapy duration, antibiotic dosages, and administration routes for mitigating negative effects of antibiotic therapies. Furthermore, this review highlights that studies on aminoglycosides are almost inexistent, and they should be increased, considering that aminoglycosides are the first most commonly used antibiotic family in companion animals. Harmonization of experimental procedures is necessary in this research field. In fact, current studies are based on different experimental set-up varying for antibiotic dosage, regimen, administration, and downstream microbiota analysis. In the future, shotgun metagenomics coupled with long-reads sequencing should become a standard experimental approach enabling to gather comprehensive knowledge on GIM in terms of composition and taxonomic functions, and of ARGs. Decorticating GIM in animals will unveil revolutionary strategies for medication and improvement of animals' health status, with positive consequences on global health.
Collapse
|
21
|
Hung YP, Lee CC, Lee JC, Tsai PJ, Hsueh PR, Ko WC. The Potential of Probiotics to Eradicate Gut Carriage of Pathogenic or Antimicrobial-Resistant Enterobacterales. Antibiotics (Basel) 2021; 10:antibiotics10091086. [PMID: 34572668 PMCID: PMC8470257 DOI: 10.3390/antibiotics10091086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/31/2022] Open
Abstract
Probiotic supplements have been used to decrease the gut carriage of antimicrobial-resistant Enterobacterales through changes in the microbiota and metabolomes, nutrition competition, and the secretion of antimicrobial proteins. Many probiotics have shown Enterobacterales-inhibiting effects ex vivo and in vivo. In livestock, probiotics have been widely used to eradicate colon or environmental antimicrobial-resistant Enterobacterales colonization with promising efficacy for many years by oral supplementation, in ovo use, or as environmental disinfectants. In humans, probiotics have been used as oral supplements for infants to decease potential gut pathogenic Enterobacterales, and probiotic mixtures, especially, have exhibited positive results. In contrast to the beneficial effects in infants, for adults, probiotic supplements might decrease potentially pathogenic Enterobacterales, but they fail to completely eradicate them in the gut. However, there are several ways to improve the effects of probiotics, including the discovery of probiotics with gut-protection ability and antimicrobial effects, the modification of delivery methods, and the discovery of engineered probiotics. The search for multifunctional probiotics and synbiotics could render the eradication of “bad” Enterobacterales in the human gut via probiotic administration achievable in the future.
Collapse
Affiliation(s)
- Yuan-Pin Hung
- Department of Internal Medicine, Tainan Hospital, Ministry of Health and Welfare, Tainan 700, Taiwan;
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
| | - Ching-Chi Lee
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
- Clinical Medicine Research Center, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan
| | - Jen-Chieh Lee
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan;
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan
- Department of Pathology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan
| | - Po-Ren Hsueh
- Departments of Laboratory Medicine and Internal Medicine, China Medical University Hospital, School of Medicine, China Medical University, Taichung 404, Taiwan
- Correspondence: (P.-R.H.); (W.-C.K.)
| | - Wen-Chien Ko
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan
- Correspondence: (P.-R.H.); (W.-C.K.)
| |
Collapse
|
22
|
Verma S, Dutta SK, Firnberg E, Phillips L, Vinayek R, Nair PP. Identification and engraftment of new bacterial strains by shotgun metagenomic sequence analysis in patients with recurrent Clostridioides difficile infection before and after fecal microbiota transplantation and in healthy human subjects. PLoS One 2021; 16:e0251590. [PMID: 34252073 PMCID: PMC8274925 DOI: 10.1371/journal.pone.0251590] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 04/29/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Recurrent Clostridioides diffícile infection (RCDI) is associated with major bacterial dysbiosis and colitis. Fecal microbiota transplantation (FMT) is a highly effective therapeutic modality for RCDI. While several studies have identified bacterial species associated with resolution of symptoms in patients, characterization of the fecal microbiome at the bacterial strain level in RCDI patients before and after FMT and healthy donors, has been lacking. The aim of this study was to examine the ability of bacterial strains from healthy donors to engraft in the gastrointestinal tract of patients with RCDI following FMT. METHODS Fecal samples were collected from 22 patients with RCDI before and after FMT and their corresponding healthy donors. Total DNA was extracted from each sample and analyzed by shotgun metagenomic sequencing. The Cosmos-ID analysis platform was used for taxonomic assignment of sequences and calculation of the relative abundance (RA) of bacterial species and strains. From these data, the total number of bacterial strains (BSI), Shannon diversity index, dysbiosis index (DI), and bacterial engraftment factor, were calculated for each strain. FINDINGS A marked reduction (p<0·0001) in the RA of total and specific bacterial strains, especially from phylum Firmicutes, was observed in RCDI patients prior to FMT. This change was associated with an increase in the DI (p<0·0001) and in pathobiont bacterial strains from phylum Proteobacteria, such as Escherichia coli O157:H7 and Klebsiella pneumoniae UCI 34. BSI was significantly lower in this group of patients as compared to healthy donors and correlated with the Shannon Index. (p<0·0001). Identification and engraftment of bacterial strains from healthy donors revealed a greater diversity and higher relative abundance of short-chain fatty acid (SCFA)-producing bacterial strains, including Lachnospiraceae bacterium 5_1_63FAA_u_t, Dorea formicigenerans ATCC 27755, Anaerostipes hadrusand others, in RCDI patients after FMT. INTERPRETATION These observations identify a group of SCFA-producing bacterial strains from healthy donors that engraft well in patients with RCDI following FMT and are associated with complete resolution of clinical symptoms and bacterial dysbiosis.
Collapse
Affiliation(s)
- Sandeep Verma
- Division of Gastroenterology, Sinai Hospital, Baltimore MD, United States of America
| | - Sudhir K. Dutta
- Division of Gastroenterology, Sinai Hospital, Baltimore MD, United States of America
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Elad Firnberg
- Division of Gastroenterology, Sinai Hospital, Baltimore MD, United States of America
| | - Laila Phillips
- Division of Gastroenterology, Sinai Hospital, Baltimore MD, United States of America
| | - Rakesh Vinayek
- Division of Gastroenterology, Sinai Hospital, Baltimore MD, United States of America
| | - Padmanabhan P. Nair
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
- Noninvasive Technologies, Elkridge, MD, United States of America
| |
Collapse
|
23
|
Chairuk P, Zaman RU, Naphatthalung J, Jansakul C. Effect of consumption of whole egg and egg fractions on cardiovascular disease factors in adult rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:3942-3951. [PMID: 33348458 DOI: 10.1002/jsfa.11034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/23/2020] [Accepted: 12/21/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND While eggs are a low-cost source of protein, rich in macro- and micronutrients, the association of egg intake and cardiovascular disease (CVD) remains controversial. This study investigated the effect of egg consumption on CVD parameters. Eggs were boiled, separated into four fractions (whole egg, 50% yolk-reduced whole egg, egg yolk and egg white) and then freeze-dried. The different egg fractions or distilled water (control) were orally gavaged to adult male Wistar rats at 1 g kg-1 rat body weight, each day for 8 weeks, following which basal blood pressure, heart rate, complete blood cell count, blood biochemistry, body fat and liver cell lipid accumulation were determined. The vascular functions of isolated thoracic aorta were studied using classical pharmacological techniques. RESULTS In comparison to the control group, none of the egg fractions affected body weight, food intake, plasma glucose or lipid profile. The yolk group experienced increased plasma alkaline phosphatase and creatinine levels, while egg white caused decreased plasma cholesterol and blood urea nitrogen. Whole egg and egg yolk increased blood pressure and mean hemoglobin concentration and the yolk increased liver lipid accumulation. Egg white decreased the white blood cell count and body fat lipids. No changes were found in basal heart rate or vascular functions in any of the groups. CONCLUSIONS Consumption of whole egg or egg yolk at the dosage given caused hypertension, with impairment of liver and kidney functions following the intake of yolk alone. However, egg white is beneficial for the cardiovascular system as it decreased plasma cholesterol and body fat accumulation. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Pilaipan Chairuk
- Faculty of Traditional Thai Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Raihan Uz Zaman
- Faculty of Traditional Thai Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Jomkarn Naphatthalung
- Faculty of Traditional Thai Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Chaweewan Jansakul
- Faculty of Traditional Thai Medicine, Prince of Songkla University, Hat Yai, Thailand
| |
Collapse
|
24
|
Li C, Zhou W, Li M, Shu X, Zhang L, Ji G. Salvia-Nelumbinis naturalis extract protects mice against MCD diet-induced steatohepatitis via activation of colonic FXR-FGF15 pathway. Biomed Pharmacother 2021; 139:111587. [PMID: 33865013 DOI: 10.1016/j.biopha.2021.111587] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/15/2022] Open
Abstract
Salvia-Nelumbinis naturalis (SNN) formula is a traditional Chinese medicine prescription, and has been confirmed to be effective in treating non-alcoholic steatohepatitis (NASH), but the underlying mechanisms are still unknown. Here we showed that 4-week SNN administration alleviated methionine-choline-deficiency (MCD) diet-induced hepatic steatosis and inflammation as well as serum levels of alanine transaminase (ALT) increase in C57BL/6 mice. Fecal 16S rDNA sequencing indicated that SNN altered the structure of gut microbiota and partially reversed the gut dysbiosis. Simultaneously, we analyzed the fecal BA profile using liquid chromatography coupled with triple quadrupole mass spectrometry (UPLC-TQMS) -based metabolomics, and found that SNN modulated fecal BA profile, predominantly increased the microbiomes related BA species (e.g. nordeoxycholic acid) which in turn, activated farnesoid X receptor (FXR)-fibroblast growth factor 15 (FGF15) signaling pathway in the colon but not the ileum. The activation of intestinal FXR-FGF15 signaling was accompanied by increase of liver protein kinase B (PKB/Akt) phosphorylation, and decrease of p-65 subunit of NF-κB phosphorylation, resulting in less liver CD68 positive macrophages, and inflammatory cytokine IL-1β and TNF-α expression. Our results established the link between SNN treatment, gut microbiota, BA profile and NASH, which might shed light into the mechanisms behind the beneficial effects of SNN on NASH, thus provide evidence for the clinical application of SNN.
Collapse
Affiliation(s)
- Chunlin Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Meng Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xiangbing Shu
- Department of Geratology, Baoshan Branch of Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201999, China
| | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
25
|
Hashimoto-Hill S, Alenghat T. Inflammation-Associated Microbiota Composition Across Domestic Animals. Front Genet 2021; 12:649599. [PMID: 34239536 PMCID: PMC8257562 DOI: 10.3389/fgene.2021.649599] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/27/2021] [Indexed: 12/26/2022] Open
Abstract
Domestic animals represent important resources for understanding shared mechanisms underlying complex natural diseases that arise due to both genetic and environmental factors. Intestinal inflammation, particularly inflammatory bowel disease (IBD), is a significant health challenge in humans and domestic animals. While the etiology of IBD is multifactorial, imbalance of symbiotic gut microbiota has been hypothesized to play a central role in disease pathophysiology. Advances in genomic sequencing and analytical pipelines have enabled researchers to decipher the composition of the intestinal microbiota during health and in the context of naturally occurring diseases. This review compiles microbiome genomic data across domestic species and highlights a common occurrence of gut microbiome dysbiosis during idiopathic intestinal inflammation in multiple species, including dogs, cats, horses, cows, and pigs. Current microbiome data obtained from animals with intestinal inflammation are mostly limited to taxonomical analyses in association with broad clinical phenotype. In general, a pathogen or pathosymbiont were not detected. Rather, functional potential of the altered microbiota has been suggested to be one of the key etiologic factors. Among the domestic species studied, canine analyses are currently the most advanced with incorporation of functional profiling of microbiota. Canine IBD parallels features of the disease in humans, thus canines represent a strong natural model for human IBD. While deeper analyses of metagenomic data, coupled with host molecular analyses are needed, comparative studies across domestic species can reveal shared microbial alterations and regulatory mechanisms that will improve our understanding of intestinal inflammation in both animals and humans.
Collapse
Affiliation(s)
| | - Theresa Alenghat
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
26
|
Whittemore JC, Price JM, Moyers T, Suchodolski JS. Effects of Synbiotics on the Fecal Microbiome and Metabolomic Profiles of Healthy Research Dogs Administered Antibiotics: A Randomized, Controlled Trial. Front Vet Sci 2021; 8:665713. [PMID: 34124225 PMCID: PMC8187564 DOI: 10.3389/fvets.2021.665713] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/19/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Antibiotic-associated gastrointestinal signs occurred in 100% of dogs administered enrofloxacin with metronidazole in a previous study, and signs partially were mitigated by synbiotics. The objective of this randomized, double-blinded, placebo-controlled trial was to compare the fecal microbiome and metabolome of dogs administered enrofloxacin and metronidazole, followed by either a placebo or a bacterial/yeast synbiotic combination. Methods: Twenty-two healthy research dogs were randomized to two treatment groups. There were three study periods: baseline, treatment, and washout. Dogs were administered enrofloxacin (10 mg/kg qd) and metronidazole (12.5 mg/kg BID), followed 1 h later by placebo or a commercially-available synbiotic combination (BID), per os for 21 days with reevaluation 56 days thereafter. Fecal samples were collected on days 5–7 (baseline), 26–28, and 82–84. The fecal microbiome was analyzed by qPCR and sequencing of 16S rRNA genes; time-of-flight mass spectrometry was used to determine metabolomic profiles. Split plot repeated measures mixed model ANOVA was used to compare results between treatment groups. P < 0.05 was considered significant, with Benjamini and Hochberg's False Discovery Rate used to adjust for multiple comparisons. Results: Alpha diversity metrics differed significantly over time in both treatment groups, with incomplete recovery by days 82–84. Beta diversity and the dysbiosis index differed significantly over time and between treatment groups, with incomplete recovery at days 82–84 for dogs in the placebo group. Significant group-by-time interactions were noted for 15 genera, including Adlercreutzia, Bifidobacterium, Slackia, Turicibacter, Clostridium (including C. hiranonis) [Ruminococcus], Erysipelotrichaceae_g_, [Eubacterium], and Succinivibrionaceae_g_. Concurrent group and time effects were present for six genera, including Collinsella, Ruminococcaceae_g_, and Prevotella. Metabolite profiles differed significantly by group-by-time, group, and time for 28, 20, and 192 metabolites, respectively. These included short-chain fatty acid, bile acid, tryptophan, sphingolipid, benzoic acid, and cinnaminic acid metabolites, as well as fucose and ethanolamine. Changes in many taxa and metabolites persisted through days 82–84. Conclusion: Antibiotic administration causes sustained dysbiosis and dysmetabolism in dogs. Significant group-by-time interactions were noted for a number of taxa and metabolites, potentially contributing to decreased antibiotic-induced gastrointestinal effects in dogs administered synbiotics.
Collapse
Affiliation(s)
- Jacqueline C Whittemore
- Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| | - Joshua M Price
- Office of Information Technology, University of Tennessee, Knoxville, TN, United States
| | - Tamberlyn Moyers
- Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| | - Jan S Suchodolski
- The Gastrointestinal Laboratory, Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
27
|
Determining Gut Microbial Dysbiosis: a Review of Applied Indexes for Assessment of Intestinal Microbiota Imbalances. Appl Environ Microbiol 2021; 87:AEM.00395-21. [PMID: 33741632 PMCID: PMC8208139 DOI: 10.1128/aem.00395-21] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Assessing “dysbiosis” in intestinal microbial communities is increasingly considered a routine analysis in microbiota studies, and it has added relevant information to the prediction and characterization of diseases and other adverse conditions. However, dysbiosis is not a well-defined condition. Assessing “dysbiosis” in intestinal microbial communities is increasingly considered a routine analysis in microbiota studies, and it has added relevant information to the prediction and characterization of diseases and other adverse conditions. However, dysbiosis is not a well-defined condition. A variety of different dysbiosis indexes have been suggested and applied, but their underlying methodologies, as well as the cohorts and conditions for which they have been developed, differ considerably. To date, no comprehensive overview and comparison of all the different methodologies and applications of such indexes is available. Here, we list all types of dysbiosis indexes identified in the literature, introduce their methodology, group them into categories, and discuss their potential descriptive and clinical applications as well as their limitations. Thus, our focus is not on the implications of dysbiosis for disease but on the methodological approaches available to determine and quantify this condition.
Collapse
|
28
|
Abstract
Probiotics/or synbiotics products for small animals do not fulfill the criteria required to qualify as a probiotic. Studies explaining modes of action are lacking. Outcome measures are inconsistent, with some trials assessing only nonspecific routine diagnostic parameters or fecal scores. Preliminary evidence shows that specific preparations are beneficial in parvovirus infections and acute hemorrhagic diarrhea syndrome in dogs and in Tritrichomonas fetus infection in cats. In dogs, inflammatory bowel disease specific probiotics can decrease clinical severity. More studies focusing on functional outcomes in dogs and cats with well-defined diseases to allow evidence-based clinical use of probiotics and synbiotics are needed.
Collapse
Affiliation(s)
- Silke Salavati Schmitz
- Hospital for Small Animals, Royal (Dick) School of Veterinary Studies, The Roslin Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK.
| |
Collapse
|
29
|
Scarsella E, Segato J, Zuccaccia D, Swanson KS, Stefanon B. An application of nuclear magnetic resonance spectroscopy to study faecal canine metabolome. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1925602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Elisa Scarsella
- Dipartimento di Scienze Agroalimentari, Ambientali e Animali, Università di Udine, Udine, Italy
| | - Jacopo Segato
- Dipartimento di Scienze Agroalimentari, Ambientali e Animali, Università di Udine, Udine, Italy
| | - Daniele Zuccaccia
- Dipartimento di Scienze Agroalimentari, Ambientali e Animali, Università di Udine, Udine, Italy
| | - Kelly S. Swanson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bruno Stefanon
- Dipartimento di Scienze Agroalimentari, Ambientali e Animali, Università di Udine, Udine, Italy
| |
Collapse
|
30
|
Pet-Human Gut Microbiome Host Classifier Using Data from Different Studies. Microorganisms 2020; 8:microorganisms8101591. [PMID: 33076521 PMCID: PMC7602744 DOI: 10.3390/microorganisms8101591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/30/2022] Open
Abstract
(1) Background: microbiome host classification can be used to identify sources of contamination in environmental data. However, there is no ready-to-use host classifier. Here, we aimed to build a model that would be able to discriminate between pet and human microbiomes samples. The challenge of the study was to build a classifier using data solely from publicly available studies that normally contain sequencing data for only one type of host. (2) Results: we have developed a random forest model that distinguishes human microbiota from domestic pet microbiota (cats and dogs) with 97% accuracy. In order to prevent overfitting, samples from several (at least four) different projects were necessary. Feature importance analysis revealed that the model relied on several taxa known to be key components in domestic cat and dog microbiomes (such as Fusobacteriaceae and Peptostreptococcaeae), as well as on some taxa exclusively found in humans (as Akkermansiaceae). (3) Conclusion: we have shown that it is possible to make a reliable pet/human gut microbiome classifier on the basis of the data collected from different studies.
Collapse
|
31
|
Radwan S, Gilfillan D, Eklund B, Radwan HM, El Menofy NG, Lee J, Kapuscinski M, Abdo Z. A comparative study of the gut microbiome in Egyptian patients with Type I and Type II diabetes. PLoS One 2020; 15:e0238764. [PMID: 32903276 PMCID: PMC7480833 DOI: 10.1371/journal.pone.0238764] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Diabetes remains a growing public health concern in Egypt, as prevalence of Type II diabetes (TIID) has nearly tripled there in the last two decades. Egypt was ranked ninth worldwide in number of diabetes cases, with prevalence of 15.56% among adults. Recent studies have proposed that disturbance of gut microbiota could influence TIID development and indicated associations between a reduced diversity in microbiomes and Type I diabetes (TID). In the present study, we investigated the composition and abundance of the bacterial microbiome in disease state (TID and TIID) of Egyptian patients. Our goal in this study was to characterize features of the gut microbiota and possible differences associated with TID and TIID in this population. METHODS DNA was extracted from fecal samples taken from 22 TID and 18 TIID outpatients of Al-Hussein hospital, Cairo, Egypt. 16S rRNA amplicon sequencing was used to characterize the bacterial taxa and these reads were processed using the software mothur with analysis utilizing packages vegan, phyloseq and metagenomSeq in R. RESULTS AND CONCLUSIONS Our results highlighted a significant increase in abundance of Gram negative, potentially opportunistic pathogenic taxa (Pseudomonas, Prevotella) in all diabetic groups, compared to the control. Lipopolysccharide (LPS), a component of the gram-negative bacterial wall, can activate local immune response and may result in low-grade systemic inflammation contributing to insulin resistance. The gram-positive Gemella, which is associated with increased risk to diabetes, also had a significant increase in abundance in all diabetic groups, compared to the control. In contrast, the commensal bacterial taxa Turicibacter, Terrisporobacter and Clostridium were found to be more abundant in the control group than in TID. Further studies are needed to understand the role of these taxa in health and disease. Lower Richness and low Shannon diversity, though not statistically significant, were observed for TID subjects with no glucose control and with onset of liver disease or hypertension compared to other subjects. In addition, large variation in alpha diversity within the control group could also be observed. Future studies will include larger samples sizes to further elucidate these findings, as well as possible metagenomic studies to examine the intriguing function of significant microbes.
Collapse
Affiliation(s)
- Sahar Radwan
- Microbiology and Immunology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Darby Gilfillan
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Bridget Eklund
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Hend M. Radwan
- Microbiology and Immunology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Nagwan G. El Menofy
- Microbiology and Immunology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Justin Lee
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Marylee Kapuscinski
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Zaid Abdo
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| |
Collapse
|
32
|
Lyu Y, Su C, Verbrugghe A, Van de Wiele T, Martos Martinez-Caja A, Hesta M. Past, Present, and Future of Gastrointestinal Microbiota Research in Cats. Front Microbiol 2020; 11:1661. [PMID: 32793152 PMCID: PMC7393142 DOI: 10.3389/fmicb.2020.01661] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/25/2020] [Indexed: 12/17/2022] Open
Abstract
The relationship between microbial community and host has profound effects on the health of animals. A balanced gastrointestinal (GI) microbial population provides nutritional and metabolic benefits to its host, regulates the immune system and various signaling molecules, protects the intestine from pathogen invasion, and promotes a healthy intestinal structure and an optimal intestinal function. With the fast development of next-generation sequencing, molecular techniques have become standard tools for microbiota research, having been used to demonstrate the complex intestinal ecosystem. Similarly to other mammals, the vast majority of GI microbiota in cats (over 99%) is composed of the predominant bacterial phyla Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria. Many nutritional and clinical studies have shown that cats' microbiota can be affected by several different factors including body condition, age, diet, and inflammatory diseases. All these factors have different size effects, and some of these may be very minor, and it is currently unknown how important these are. Further research is needed to determine the functional variations in the microbiome in disease states and in response to environmental and/or dietary modulations. Additionally, further studies are also needed to explain the intricate relationship between GI microbiota and the genetics and immunity of its host. This review summarizes past and present knowledge of the feline GI microbiota and looks into the future possibilities and challenges of the field.
Collapse
Affiliation(s)
- Yang Lyu
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Chunxia Su
- Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Adronie Verbrugghe
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Tom Van de Wiele
- Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Ana Martos Martinez-Caja
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Myriam Hesta
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
33
|
Garcia-Mazcorro JF, Minamoto Y, Kawas JR, Suchodolski JS, de Vos WM. Akkermansia and Microbial Degradation of Mucus in Cats and Dogs: Implications to the Growing Worldwide Epidemic of Pet Obesity. Vet Sci 2020; 7:vetsci7020044. [PMID: 32326394 PMCID: PMC7355976 DOI: 10.3390/vetsci7020044] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 02/07/2023] Open
Abstract
Akkermansia muciniphila is a mucin-degrading bacterium that has shown the potential to provide anti-inflammatory and anti-obesity effects in mouse and man. We here focus on companion animals, specifically cats and dogs, and evaluate the microbial degradation of mucus and its health impact in the context of the worldwide epidemic of pet obesity. A literature survey revealed that the two presently known Akkermansia spp., A. muciniphila and A. glycaniphila, as well as other members of the phylum of Verrucomicrobia seem to be neither very prevalent nor abundant in the digestive tract of cats and dog. While this may be due to methodological aspects, it suggests that bacteria related to Akkermansia are not the major mucus degraders in these pets and hence other mucus-utilizing taxa may deserve attention. Hence, we will discuss the potential of these endogenous mucus utilizers and dietary interventions to boost these as well as the use of Akkermansia spp. related bacteria or their components as strategies to target feline and canine obesity.
Collapse
Affiliation(s)
- Jose F. Garcia-Mazcorro
- Research and Development, MNA de Mexico, San Nicolas de los Garza, Nuevo Leon 66477, Mexico
- Correspondence: ; Tel.: +52-81-8850-5204
| | | | - Jorge R. Kawas
- Faculty of Agronomy, Universidad Autonoma de Nuevo Leon, General Escobedo, Nuevo Leon 66050, Mexico;
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4474, USA;
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, 6708 WE Wageningen, The Netherlands;
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, P.O. Box 63, 00014 Helsinki, Finland
| |
Collapse
|
34
|
Wu YHS, Lin YL, Huang C, Chiu CH, Nakthong S, Chen YC. Cardiac protection of functional chicken-liver hydrolysates on the high-fat diet induced cardio-renal damages via sustaining autophagy homeostasis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:2443-2452. [PMID: 31951016 DOI: 10.1002/jsfa.10261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/02/2020] [Accepted: 01/10/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Cardio-renal syndrome (CRS) is an integrative problem related to chronic malnutrition, obesity, etc. Amino acids and peptides are regarded as protective and essential for tissues. Pepsin-digested chicken liver hydrolysates (CLHs), which are made from the byproducts of the poultry industry, are amino-acid based and of animal origin, and may be protective against the myocardial and renal damage induced by a high-fat diet (HFD). RESULTS Our results showed that CLHs contain large quantities of anserine, taurine, and branched-chain amino acids (BCAAs), and supplementing the diet with CLHs reduced (P < 0.05) weight gain, liver weight, peri-renal fat mass / adipocyte-area sizes, serum total cholesterol (TC), aspartate aminotransferase (AST), and low-density lipoprotein cholesterol (LDLC) levels in HFD-fed mice but increased (P < 0.05) serum high-density lipoprotein cholesterol (HDLC) levels. By histological analyses, CLHs alleviated (P < 0.05) renal lipid deposition and fibrosis, as well as cardiac fibrosis and inflammation of HFD-fed mice. Meanwhile, increased (P < 0.05) inflammatory and fibrotic cytokines levels in the myocardia of the HFD-fed mice were downregulated (P < 0.05) by CLH supplementation. Regarding autophagy-related protein levels, protective effects of CLHs on the myocardia against HFD feeding may result from the early blockade of the autophagy pathway to prevent autophagosome accumulation. CONCLUSION Functional CLHs could be a novel food ingredient as a cardio-renal protective agent against a high-fat dietary habit in a niche market. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yi-Hsieng Samuel Wu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yi-Ling Lin
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Chien Huang
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Chih-Hsien Chiu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Sasitorn Nakthong
- Department of Animal Science, Faculty of Agriculture at Kamphaeng Saen, Kasetsart University, Nakhon Pathom, Thailand
| | - Yi-Chen Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|