1
|
Li W, Yang J. Investigating the Anna Karenina principle of the breast microbiome. BMC Microbiol 2025; 25:81. [PMID: 39979818 PMCID: PMC11841003 DOI: 10.1186/s12866-024-03738-y] [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: 06/06/2024] [Accepted: 12/30/2024] [Indexed: 02/22/2025] Open
Abstract
The relationship between the microbiome and disease has long been a central focus of research in human microbiome. Inspired by Leo Tolstoy's dictum, the Anna Karenina Principle (AKP) offers a framework for understanding the complex dynamics of microbial communities in response to perturbations, suggesting that dysbiotic individuals exhibit greater variability/heterogeneity in their microbiome compared to healthy counterparts. While some studies have proved the alignment of microbiome responses to disease with the AKP effect, it remains uncertain whether the human breast microbiome responds similarly to breast disease. This study used beta-diversity and similarity in Hill numbers, along with shared species analysis (SSA), to explore this issue. We observed that during mastitis, changes in both the taxa richness and composition in the breast milk microbiome align with the AKP effect, while alterations in abundant taxa exhibit an anti-AKP effect. The response of breast tissue microbiome to breast cancer differs from that of milk microbiome to mastitis. Breast cancer induce anti-AKP effects in taxa richness, and non-AKP effects in common taxa and taxa composition. Overall, our findings identified different responses to breast diseases across taxa abundance in the breast microbiome. Mastitis primarily involves increasing the heterogeneity of rare taxa in the breast milk microbiome, while breast cancer associates with decreased dispersion of rare taxa in the tissue microbiome.
Collapse
Affiliation(s)
- Wendy Li
- College of Biological Sciences and Technology, Taiyuan Normal University, Jinzhong, 030600, China.
| | - Jinghui Yang
- Department of Pediatrics, The First People's Hospital of Yunnan Province, Kunming, 650032, China.
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China.
- Yunnan Province Clinical Research Center for Hematologic Disease, Kunming, 650032, China.
- Yunnan Province Clinical Center for Hematologic Disease, Kunming, 650032, China.
| |
Collapse
|
2
|
Reuben RC, Torres C. Integrating the milk microbiome signatures in mastitis: milk-omics and functional implications. World J Microbiol Biotechnol 2025; 41:41. [PMID: 39826029 PMCID: PMC11742929 DOI: 10.1007/s11274-024-04242-1] [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: 10/13/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025]
Abstract
Mammalian milk contains a variety of complex bioactive and nutritional components and microorganisms. These microorganisms have diverse compositions and functional roles that impact host health and disease pathophysiology, especially mastitis. The advent and use of high throughput omics technologies, including metagenomics, metatranscriptomics, metaproteomics, metametabolomics, as well as culturomics in milk microbiome studies suggest strong relationships between host phenotype and milk microbiome signatures in mastitis. While single omics studies have undoubtedly contributed to our current understanding of milk microbiome and mastitis, they often provide limited information, targeting only a single biological viewpoint which is insufficient to provide system-wide information necessary for elucidating the biological footprints and molecular mechanisms driving mastitis and milk microbiome dysbiosis. Therefore, integrating a multi-omics approach in milk microbiome research could generate new knowledge, improve the current understanding of the functional and structural signatures of the milk ecosystem, and provide insights for sustainable mastitis control and microbiome management.
Collapse
Affiliation(s)
- Rine Christopher Reuben
- Biology Department, King's College, 133 North River Street, Wilkes-Barre, PA, 18711, USA.
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006, Logroño, Spain.
| | - Carmen Torres
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006, Logroño, Spain
| |
Collapse
|
3
|
Castro-Navarro I, Pace RM, Williams JE, Pace CDW, Kaur H, Piaskowski J, Aragón A, Rodríguez JM, McGuire MA, Fernandez L, McGuire MK. Immunological composition of human milk before and during subclinical and clinical mastitis. Front Immunol 2025; 15:1532432. [PMID: 39896819 PMCID: PMC11782115 DOI: 10.3389/fimmu.2024.1532432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Mastitis, an inflammatory condition affecting more than 25% of breastfeeding women, is usually associated with reduced milk secretion, pain, and discomfort, which often leads to early cessation of breastfeeding. Although the etiology of mastitis is multifactorial, a pro-inflammatory state of the mammary gland might be a risk factor. However, changes in milk composition, and specifically in the milk immune profile, prior to and during mastitis have not been well described. To help close this research gap, we documented the immune profiles of milk produced by both breasts of 10 women experiencing clinical (CM) and 8 women experiencing subclinical (SCM) mastitis during the week of sign/symptom development as well as the week prior and compared them with milk produced by 14 healthy controls. CM was defined as having signs/symptoms of mastitis, whereas SCM was presumed if the participant did not have signs/symptoms of CM, but her milk had a somatic cell count >400,000 cell/mL and/or sodium-to-potassium (Na/K) ratio >1.0. Concentration of 36 immune factors (including immunoglobulins, cytokines, chemokines, and growth factors) was quantified via immunoassays. Milk produced by women who developed CM had distinct immune profiles the week prior to diagnosis, particularly elevated concentrations of pro-inflammatory cytokine IL-1β and regulatory cytokines IL-2, IL-4 and IL-10. In contrast, immune profiles in milk produced by women with SCM did not differ from that produced by healthy women or those with CM the week prior to mastitis onset. Once mastitis appeared, marked changes in milk's immune profile were observed in both CM and SCM groups. CM was characterized by elevated concentrations of 27 compounds, including pro-inflammatory cytokines (IL-1β, IL-1ra, and TNFα) and chemokines (including IL-8, eotaxin, IP-10, MCP-1, MIP1α, and MIP1β), compared to healthy controls. Milk's immune profile during SCM was intermediate, showing higher levels of IL-6, IFNγ, and MCP-1 compared to healthy controls, suggesting a milder, more controlled immune response compared to CM. Only milk produced by the mastitis-affected breast had altered immune profiles. Further research is needed to determine if these differences in milk's immune profiles can be used to improve mastitis risk prediction prior to onset of symptoms.
Collapse
Affiliation(s)
- Irma Castro-Navarro
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Ryan M. Pace
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
- College of Nursing, University of South Florida, Tampa, FL, United States
- Microbiomes Institute, University of South Florida, Tampa, FL, United States
| | - Janet E. Williams
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Christina D. W. Pace
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Harpreet Kaur
- Statistical Programs, College of Agricultural and Life Sciences, University of Idaho, Moscow, ID, United States
| | - Julia Piaskowski
- Statistical Programs, College of Agricultural and Life Sciences, University of Idaho, Moscow, ID, United States
| | - Alberto Aragón
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, Madrid, Spain
- Instituto Pluridisciplinar, Complutense University of Madrid, Madrid, Spain
| | - Juan M. Rodríguez
- Instituto Pluridisciplinar, Complutense University of Madrid, Madrid, Spain
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Mark A. McGuire
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Leonides Fernandez
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, Madrid, Spain
- Instituto Pluridisciplinar, Complutense University of Madrid, Madrid, Spain
| | - Michelle K. McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| |
Collapse
|
4
|
Zhou Y, Jiang M, Li X, Shen K, Zong H, Lv Q, Shen B. Bibliometric and visual analysis of human microbiome-breast cancer interactions: current insights and future directions. Front Microbiol 2024; 15:1490007. [PMID: 39717276 PMCID: PMC11664440 DOI: 10.3389/fmicb.2024.1490007] [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: 09/02/2024] [Accepted: 11/20/2024] [Indexed: 12/25/2024] Open
Abstract
The composition of the gut microbiome differs from that of healthy individuals and is closely linked to the progression and development of breast cancer. Recent studies have increasingly examined the relationship between microbial communities and breast cancer. This study analyzed the research landscape of microbiome and breast cancer, focusing on 736 qualified publications from the Web of Science Core Collection (WoSCC). Publications in this field are on the rise, with the United States leading in contributions, followed by China and Italy. Despite this strong output, the centrality value of China in this field is comparatively low at ninth, highlighting a gap between the quantity of research and its global impact. This pattern is repetitively observed in institutional contributions, with a predominance of Western institutes among the top contributors, underscoring a potential research quality gap in China. Keyword analysis reveals that research hotspots are focused on the effect of microbiome on breast cancer pathogenesis and tumor metabolism, with risk factors and metabolic pathways being the most interesting areas. Publications point to a shift toward anti-tumor therapies and personalized medicine, with clusters such as "anti-tumor" and "potential regulatory agent" gaining prominence. Additionally, intratumor bacteria studies have emerged as a new area of significant interest, reflecting a new direction in research. The University of Helsinki and Adlercreutz H are influential institutions and researchers in this field. Current trends in microbiome and breast cancer research indicate a significant shift toward therapeutic applications and personalized medicine. Strengthening international collaborations and focusing on research quality is crucial for advancing microbiome and breast cancer research.
Collapse
Affiliation(s)
| | | | | | | | | | - Qing Lv
- Department of Breast Surgery and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Sichuan, China
| | - Bairong Shen
- Department of Breast Surgery and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Sichuan, China
| |
Collapse
|
5
|
Dalwadi P, Nathani N, Chauhan K, Mansuri J, Koringa P, Bhatt V, Kunjadiya AP. Whole-genome sequencing of bacteria accountable for lactational mastitis in humans combined with an examination of their antibiotic resistance profiles. Braz J Microbiol 2024; 55:3827-3838. [PMID: 39320640 PMCID: PMC11711416 DOI: 10.1007/s42770-024-01519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/08/2024] [Indexed: 09/26/2024] Open
Abstract
Lactational mastitis, a common condition affecting nursing mothers, is characterized by mammary gland inflammation during lactation. This inflammatory response typically occurs due to bacterial infection. The discomfort and pain associated with lactational mastitis can significantly impact a mother's ability to breastfeed comfortably and may lead to the cessation of breastfeeding altogether if left untreated. Antibiotics are commonly prescribed to target the bacteria causing the infection and alleviate symptoms, aiming to treat the infection. Nevertheless, a notable worry linked to antibiotic use is the emergence of antibiotic resistance, compounded by the possible persistence of antibiotics in milk. Additionally, lactational mastitis is characterized by its polymicrobial nature. In this study, bacteria were isolated from infected breast milk samples and whole-genome sequencing was performed on eleven isolates to accurately identify the bacteria and assess their antibiotic resistance profiles. Using Galaxy tools and the ResFinder database, we identified Bacillus paraanthracis, Bacillus altitudinis, Staphylococcus aureus, Bacillus cereus, Escherichia coli, Alcaligenes faecalis, and Bacillus licheniformis, along with antibiotic-resistant genes like fosB1, cat86, erm (D), blaZ, and mdf (A). ABRicate aided in antimicrobial resistance (AMR) gene analysis, and CARD visualized their distribution. Our study demonstrates that the severity of infection is directly proportional to an increase in somatic cell count (SCC). This research sheds light on microbial diversity in lactational mastitis milk and provides crucial insights into antibiotic-resistance genes. Utilizing bioinformatics tools, such as those employed in this study, can inform the design of effective treatment strategies for lactational mastitis infections.
Collapse
Affiliation(s)
- Priyanka Dalwadi
- P. G. Department of Applied and Interdisciplinary Sciences, Sardar Patel University, V. V. Nagar, Gujarat, 388 120, India
| | - Neelam Nathani
- School of Applied Sciences & Technology, Gujarat Technological University, Ahmedabad, 382 424, Gujarat, India
| | - Kshipra Chauhan
- School of Applied Sciences & Technology, Gujarat Technological University, Ahmedabad, 382 424, Gujarat, India
| | - Jasmine Mansuri
- P. G. Department of Applied and Interdisciplinary Sciences, Sardar Patel University, V. V. Nagar, Gujarat, 388 120, India
| | - Prakash Koringa
- Department of Animal Biotechnology, College of Veterinary Science and Animal Husbandry, Kamdhenu University, Anand, 388 001, Gujarat, India
| | - Vaibhav Bhatt
- School of Applied Sciences & Technology, Gujarat Technological University, Ahmedabad, 382 424, Gujarat, India
| | - Anju P Kunjadiya
- P. G. Department of Applied and Interdisciplinary Sciences, Sardar Patel University, V. V. Nagar, Gujarat, 388 120, India.
| |
Collapse
|
6
|
Saleem A, Saleem Bhat S, A. Omonijo F, A Ganai N, M. Ibeagha-Awemu E, Mudasir Ahmad S. Immunotherapy in mastitis: state of knowledge, research gaps and way forward. Vet Q 2024; 44:1-23. [PMID: 38973225 PMCID: PMC11232650 DOI: 10.1080/01652176.2024.2363626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/27/2024] [Indexed: 07/09/2024] Open
Abstract
Mastitis is an inflammatory condition that affects dairy cow's mammary glands. Traditional treatment approaches with antibiotics are increasingly leading to challenging scenarios such as antimicrobial resistance. In order to mitigate the unwanted side effects of antibiotics, alternative strategies such as those that harness the host immune system response, also known as immunotherapy, have been implemented. Immunotherapy approaches to treat bovine mastitis aims to enhance the cow's immune response against pathogens by promoting pathogen clearance, and facilitating tissue repair. Various studies have demonstrated the potential of immunotherapy for reducing the incidence, duration and severity of mastitis. Nevertheless, majority of reported therapies are lacking in specificity hampering their broad application to treat mastitis. Meanwhile, advancements in mastitis immunotherapy hold great promise for the dairy industry, with potential to provide effective and sustainable alternatives to traditional antibiotic-based approaches. This review synthesizes immunotherapy strategies, their current understanding and potential future perspectives. The future perspectives should focus on the development of precision immunotherapies tailored to address individual pathogens/group of pathogens, development of combination therapies to address antimicrobial resistance, and the integration of nano- and omics technologies. By addressing research gaps, the field of mastitis immunotherapy can make significant strides in the control, treatment and prevention of mastitis, ultimately benefiting both animal and human health/welfare, and environment health.
Collapse
Affiliation(s)
- Afnan Saleem
- Division of Animal Biotechnology, SKUAST-K, Srinagar, India
| | | | - Faith A. Omonijo
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Canada
| | | | - Eveline M. Ibeagha-Awemu
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Canada
| | | |
Collapse
|
7
|
Cabrera-Rubio R, Calvo C, Alcolea S, Bergia M, Atucha J, Pozo F, Casas I, Arroyas M, Collado MC, García-García ML. Gut and respiratory tract microbiota in children younger than 12 months hospitalized for bronchiolitis compared with healthy children: can we predict the severity and medium-term respiratory outcome? Microbiol Spectr 2024; 12:e0255623. [PMID: 38785596 PMCID: PMC11218511 DOI: 10.1128/spectrum.02556-23] [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: 06/18/2023] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Growing evidence indicates that gut and respiratory microbiota have a potential key effect on bronchiolitis, mainly caused by respiratory syncytial virus (RSV). This was a prospective study of 96 infants comparing infants with bronchiolitis (n = 57, both RSV and non-RSV associated) to a control group (n = 39). Gut (feces) and respiratory [nasopharyngeal aspirate (NPA)] microbial profiles were analyzed by 16S rRNA amplicon sequencing, and respiratory viruses were identified by PCR. Clinical data of the acute episode and follow-up during the first year after infection were recorded. Pairwise comparisons showed significant differences in the gut (R2 = 0.0639, P = 0.006) and NPA (R2 = 0.0803, P = 0.006) microbiota between cases and controls. A significantly lower gut microbial richness and an increase in the NPA microbial diversity (mainly due to an increase in Haemophilus, Streptococcus, and Neisseria) were observed in the infants with bronchiolitis, in those with the most severe symptoms, and in those who subsequently developed recurrent wheezing episodes after discharge. In NPA, the higher microbial richness differed significantly between the control group and the non-RSV bronchiolitis group (P = 0.01) and between the control group and the RSV bronchiolitis group (P = 0.001). In the gut, the richness differed significantly between the control group and the non-RSV group (P = 0.01) and between the control group and the RSV bronchiolitis group (P = 0.001), with higher diversity in the RSV group. A distinct respiratory and intestinal microbial pattern was observed in infants with bronchiolitis compared with controls. The presence of RSV was a main factor for dysbiosis. Lower gut microbial richness and increased respiratory microbial diversity were associated with respiratory morbidity during follow-up. IMPORTANCE Both the intestinal and respiratory microbiota of children with bronchiolitis, especially those with respiratory syncytial virus infection, are altered and differ from that of healthy children. The microbiota pattern in the acute episode could identify those children who will later have other respiratory episodes in the first year of life. Preventive measures could be adopted for this group of infants.
Collapse
Affiliation(s)
- Raul Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Cristina Calvo
- Paediatric Infectious Diseases Department, La Paz University Hospital, Madrid, Spain
- La Paz University Hospital Institute for Health Research (IdiPAZ), Madrid, Spain
- Translational Research Network in Paediatric Infectious Diseases (RITIP), Madrid, Spain
- CIBER of Infectious Diseases (CIBERINFEC ISCIII), Madrid, Spain
| | - Sonia Alcolea
- Paediatric Infectious Diseases Department, La Paz University Hospital, Madrid, Spain
- La Paz University Hospital Institute for Health Research (IdiPAZ), Madrid, Spain
| | - María Bergia
- Paediatric Infectious Diseases Department, La Paz University Hospital, Madrid, Spain
- La Paz University Hospital Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Jorge Atucha
- Paediatric Infectious Diseases Department, La Paz University Hospital, Madrid, Spain
- La Paz University Hospital Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Francisco Pozo
- Respiratory Viruses and Influenza Unit at the National Centre for Microbiology ISCIII (CIBERESP ISCIII), Madrid, Spain
| | - Inmaculada Casas
- Respiratory Viruses and Influenza Unit at the National Centre for Microbiology ISCIII (CIBERESP ISCIII), Madrid, Spain
| | - María Arroyas
- Paediatric Department, Severo Ochoa University Hospital, Leganés, Madrid, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Maria Luz García-García
- Translational Research Network in Paediatric Infectious Diseases (RITIP), Madrid, Spain
- CIBER of Infectious Diseases (CIBERINFEC ISCIII), Madrid, Spain
- Paediatric Department, Severo Ochoa University Hospital, Leganés, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia de Arana (IDIPHISA), Madrid, Spain
| |
Collapse
|
8
|
Saifi F, Jeoboam B, Demory Beckler M, Costin JM. The Association Between Lactational Infective Mastitis and the Microbiome: Development, Onset, and Treatments. Cureus 2024; 16:e62717. [PMID: 39036221 PMCID: PMC11259407 DOI: 10.7759/cureus.62717] [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: 03/15/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
Lactational infective mastitis (LIM) was previously thought to occur due to trapped milk causing inadequate milk drainage and consequent infection. However, advances in genome sequencing techniques have shown that the abundance of Staphylococcus aureus, Staphylococcus epidermidis, Lactobacilli species, and Bifidobacterium species in the breast milk of lactating women play a key role in the development of LIM. Recent discoveries have revealed that the breast milk microbiome is composed of bacteria and other microorganisms, which are seeded through multiple pathways and are influenced by maternal factors. An imbalance in the microbial abundance in breast milk can lead to LIM. Given that this infection can cause early termination of breastfeeding, it is imperative to discuss prevention and treatment options. The objective of this review is to highlight the pathogens involved in LIM affecting human mothers, routes of bacterial transfer, and contributing factors that may influence changes in the composition of the milk microbiota, as well as propose preventative and curative treatment options.
Collapse
Affiliation(s)
- Farishta Saifi
- Biomedical Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Benscar Jeoboam
- Biomedical Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Michelle Demory Beckler
- Microbiology and Immunology, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Joshua M Costin
- Medical Education, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| |
Collapse
|
9
|
Treven P, Paveljšek D, Kostanjšek R, Golob M, Bogovič Matijašič B, Mohar Lorbeg P. In vitro model of human mammary gland microbial colonization (MAGIC) demonstrates distinctive cytokine response to imbalanced human milk microbiota. Microbiol Spectr 2024; 12:e0236923. [PMID: 38289112 PMCID: PMC10913382 DOI: 10.1128/spectrum.02369-23] [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: 06/06/2023] [Accepted: 12/19/2023] [Indexed: 03/06/2024] Open
Abstract
Despite the established concept of the human mammary gland (MG) as a habitat with its own microbiota, the exact mechanism of MG colonization is still elusive and a well-characterized in vitro model would reinforce studies of the MG microbiota development. We aimed to establish and characterize an in vitro cell model for studying MAmmary Gland mIcrobial Colonization (MAGIC) model. We used the immortalized cell line MCF10A, which expresses the strong polarized phenotype similar to MG ductal epithelium when cultured on a permeable support (Transwell). We analyzed the surface properties of the MAGIC model by gene expression analysis of E-cadherin, tight junction proteins, and mucins and by scanning electron microscopy. To demonstrate the applicability of the model, we tested the adhesion capability of the whole human milk (HM) microbial community and the cellular response of the model when challenged directly with raw HM samples. MCF10A on permeable supports differentiated and formed a tight barrier, by upregulation of CLDN8, MUC1, MUC4, and MUC20 genes. The surface of the model was covered with mucins and morphologically diverse with at least two cell types and two types of microvilli. Cells in the MAGIC model withstood the challenge with heat-treated HM samples and responded differently to the imbalanced HM microbiota by distinctive cytokine response. The microbial profile of the bacteria adhered on the MAGIC model reflected the microbiological profile of the input HM samples. The well-studied MAGIC model could be useful for studies of bacterial attachment to the MG and for in vitro studies of biofilm formation and microbiota development.IMPORTANCEThe MAGIC model may be particularly useful for studies of bacterial attachment to the surface of the mammary ducts and for in vitro studies of biofilm formation and the development of the human mammary gland (MG) microbiota. The model is also useful for immunological studies of the interaction between bacteria and MG cells. We obtained pioneering information on which of the bacteria present in the raw human milk (HM) were able to attach to the epithelium treated directly with raw HM, as well as on the effects of bacteria on the MG epithelial cells. The MAGIC cell model also offers new opportunities for research in other areas of MG physiology, such as the effects of bioactive milk components on microbial colonization of the MG, mastitis prevention, and studies of probiotic development. Since resident MG bacteria may be an important factor in breast cancer development, the MAGIC in vitro tool also offers new opportunities for cancer research.
Collapse
Affiliation(s)
- Primož Treven
- Department of Animal Science, University of Ljubljana, Biotechnical Faculty, Institute of Dairy Science and Probiotics, Domžale, Slovenia
| | - Diana Paveljšek
- Department of Animal Science, University of Ljubljana, Biotechnical Faculty, Institute of Dairy Science and Probiotics, Domžale, Slovenia
| | - Rok Kostanjšek
- Department of Biology, University of Ljubljana, Biotechnical Faculty, Chair of Zoology, Ljubljana, Slovenia
| | - Majda Golob
- University of Ljubljana, Veterinary Faculty, Institute of Microbiology and Parasitology, Ljubljana, Slovenia
| | - Bojana Bogovič Matijašič
- Department of Animal Science, University of Ljubljana, Biotechnical Faculty, Institute of Dairy Science and Probiotics, Domžale, Slovenia
| | - Petra Mohar Lorbeg
- Department of Animal Science, University of Ljubljana, Biotechnical Faculty, Institute of Dairy Science and Probiotics, Domžale, Slovenia
| |
Collapse
|
10
|
Hoque MN, Moyna Z, Faisal GM, Das ZC. Whole-Genome Sequence of the Multidrug-Resistant Staphylococcus warneri Strain G1M1F, Isolated from Mice with Mastitis. Microbiol Resour Announc 2023; 12:e0027523. [PMID: 37093041 DOI: 10.1128/mra.00275-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023] Open
Abstract
We sequenced the genome of Staphylococcus warneri G1M1F, a multidrug-resistant strain isolated from fecal samples of mice with induced mastitis. The complete genome of G1M1F consists of one chromosome of 2,504,515 bp and two plasmid replicons of 28,679 and 8,615 bp; it comprises 32.7% GC content, with 60× genome coverage.
Collapse
Affiliation(s)
- M Nazmul Hoque
- Molecular Biology and Bioinformatics Laboratory, Department of Gynaecology, Obstetrics and Reproductive Health, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Zannatara Moyna
- Molecular Biology and Bioinformatics Laboratory, Department of Gynaecology, Obstetrics and Reproductive Health, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Golam Mahbub Faisal
- Molecular Biology and Bioinformatics Laboratory, Department of Gynaecology, Obstetrics and Reproductive Health, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Ziban Chandra Das
- Molecular Biology and Bioinformatics Laboratory, Department of Gynaecology, Obstetrics and Reproductive Health, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| |
Collapse
|
11
|
Couvillion SP, Mostoller KE, Williams JE, Pace RM, Stohel IL, Peterson HK, Nicora CD, Nakayasu ES, Webb-Robertson BJM, McGuire MA, McGuire MK, Metz TO. Interrogating the role of the milk microbiome in mastitis in the multi-omics era. Front Microbiol 2023; 14:1105675. [PMID: 36819069 PMCID: PMC9932517 DOI: 10.3389/fmicb.2023.1105675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
There is growing interest in a functional understanding of milk-associated microbiota as there is ample evidence that host-associated microbial communities play an active role in host health and phenotype. Mastitis, characterized by painful inflammation of the mammary gland, is prevalent among lactating humans and agricultural animals and is associated with significant clinical and economic consequences. The etiology of mastitis is complex and polymicrobial and correlative studies have indicated alterations in milk microbial community composition. Recent evidence is beginning to suggest that a causal relationship may exist between the milk microbiota and host phenotype in mastitis. Multi-omic approaches can be leveraged to gain a mechanistic, molecular level understanding of how the milk microbiome might modulate host physiology, thereby informing strategies to prevent and ameliorate mastitis. In this paper, we review existing studies that have utilized omics approaches to investigate the role of the milk microbiome in mastitis. We also summarize the strengths and challenges associated with the different omics techniques including metagenomics, metatranscriptomics, metaproteomics, metabolomics and lipidomics and provide perspective on the integration of multiple omics technologies for a better functional understanding of the milk microbiome.
Collapse
Affiliation(s)
- Sneha P. Couvillion
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States,*Correspondence: Sneha P. Couvillion, ✉
| | - Katie E. Mostoller
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Janet E. Williams
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Ryan M. Pace
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Izabel L. Stohel
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Haley K. Peterson
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Carrie D. Nicora
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Ernesto S. Nakayasu
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Bobbie-Jo M. Webb-Robertson
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Mark A. McGuire
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Michelle K. McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Thomas O. Metz
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States,Thomas O. Metz, ✉
| |
Collapse
|
12
|
Cortés-Macías E, Selma-Royo M, Rio-Aige K, Bäuerl C, Rodríguez-Lagunas MJ, Martínez-Costa C, Pérez-Cano FJ, Collado MC. Distinct breast milk microbiota, cytokine, and adipokine profiles are associated with infant growth at 12 months: an in vitro host-microbe interaction mechanistic approach. Food Funct 2023; 14:148-159. [PMID: 36472137 DOI: 10.1039/d2fo02060b] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Breast milk (BM) is important for adequate infant development, and it contains bioactive compounds, such as bacteria, cytokines and some adipokines which play a role in infant microbial, metabolic, and immunological maturation. However, little is known about its impact on growth and development in early life. The objective of this study was to evaluate the impact of milk microbiota, cytokine, and adipokine profiles on the risk of overweight at 12 months of life to find the possible mechanisms of host-microbe interactions. In this study, BM samples from 100 healthy women collected during 15 d after birth were included. BM microbiota was analysed by 16S rRNA gene sequencing, and cytokine and adipokine levels were measured by the Luminex approach. In addition, infant weight and length were recorded during the first 12 months and z-scores were obtained according to the WHO databases. Infants were classified as risk of overweight (ROW) and no-risk of overweight (NOROW) based on their body mass index z-score (BMIZ) and infant weight-for-length z-score (WLZ) at 12 months. In order to study host-microbe interactions, epithelial intestinal and mammary cell lines were exposed to milk microbes to assess the host response by interleukin (IL)-6 production as a potential inflammatory marker. BM was dominated by Staphylococcus and Streptococcus genera, and the most abundant cytokines were IL-6 and IL-18. Leptin levels were positively correlated with the pregestational body mass index (BMI). Higher relative abundance of the Streptococcus genus was associated with higher IL-10 and higher relative abundance of the Bifidobacterium genus was associated with lower IL-6 concentrations in milk. Infant WLZ at 12 months could be partially predicted by Streptococcus genus proportions and IL-10 and IL-18 levels in BM. BM microbiota significantly induced cytokine responses in mammary epithelial cells. Higher levels of IL-6 production were observed in mammary cells exposed to BM microbiota from mothers with ROW offspring compared to mothers with NOROW offspring. In conclusion, BM microbiota is related to the cytokine profile. IL-10 and IL-18 levels and the abundance of the Streptococcus genus could affect early infant development. Further research is needed to clarify the specific impact of BM microbiota and cytokines on infant growth and the risk of overweight.
Collapse
Affiliation(s)
- Erika Cortés-Macías
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-, National Research Council (IATA-CSIC), Valencia, Spain.
| | - Marta Selma-Royo
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-, National Research Council (IATA-CSIC), Valencia, Spain.
| | - Karla Rio-Aige
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Christine Bäuerl
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-, National Research Council (IATA-CSIC), Valencia, Spain.
| | - María José Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Cecilia Martínez-Costa
- Department of Pediatrics, School of Medicine, University of Valencia, Valencia, Spain.,Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, INCLIVA, Valencia, Spain
| | - Francisco J Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-, National Research Council (IATA-CSIC), Valencia, Spain.
| |
Collapse
|
13
|
Bankvall M, Carda-Diéguez M, Mira A, Karlsson A, Hasséus B, Karlsson R, Robledo-Sierra J. Metataxonomic and metaproteomic profiling of the oral microbiome in oral lichen planus - a pilot study. J Oral Microbiol 2022; 15:2161726. [PMID: 36605405 PMCID: PMC9809343 DOI: 10.1080/20002297.2022.2161726] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background A growing body of evidence demonstrates a different bacterial composition in the oral cavity of patients with oral lichen planus (OLP). Patients and methods Buccal swab samples were collected from affected and non-affected sites of six patients with reticular OLP and the healthy oral mucosa of six control subjects. 16S rRNA gene MiSeq sequencing and mass spectrometry-based proteomics were utilised to identify the metataxonomic and metaproteomic profiles of the oral microbiome in both groups. Results From the metataxonomic analysis, the most abundant species in the three subgroups were Streptococcus oralis and Pseudomonas aeruginosa, accounting for up to 70% of the total population. Principal Coordinates Analysis showed differential clustering of samples from the healthy and OLP groups. ANCOM-BC compositional analysis revealed multiple species (including P. aeruginosa and several species of Veillonella, Prevotella, Streptococcus and Neisseria) significantly over-represented in the control group and several (including Granulicatella elegans, Gemella haemolysans and G. parahaemolysans) in patients with OLP. The metaproteomic data were generally congruent and revealed that several Gemella haemolysans-belonging peptidases and other proteins with inflammatory and virulence potential were present in OLP lesions. Conclusion Our data suggest that several bacterial species are associated with OLP. Future studies with larger cohorts should be conducted to determine their role in the aetiology of OLP and evaluate their potential as disease biomarkers.
Collapse
Affiliation(s)
- Maria Bankvall
- Department of Dental Medicine, Karolinska Institute, Huddinge, Sweden
| | - Miguel Carda-Diéguez
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| | - Alex Mira
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain,School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | | | - Bengt Hasséus
- Department of Oral Medicine and Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Roger Karlsson
- Clinical microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jairo Robledo-Sierra
- Nanoxis Consulting AB, Gothenburg, Sweden,Faculty of Dentistry, CES University, Medellin, Colombia,CONTACT Jairo Robledo-Sierra Faculty of Dentistry, CES University, Medellin, Colombia
| |
Collapse
|
14
|
Perrella SL, Anderton-May EL, McLoughlin G, Lai CT, Simmer KN, Geddes DT. Human Milk Sodium and Potassium as Markers of Mastitis in Mothers of Preterm Infants. Breastfeed Med 2022; 17:1003-1010. [PMID: 36378839 DOI: 10.1089/bfm.2022.0198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Introduction: This prospective longitudinal study examined changes in milk sodium concentration (Na) and sodium:potassium ratio (Na:K), microbiological culture, milk production, and breast health in relation to mastitis after preterm birth. Methods: We studied women who gave birth at 29-34 weeks of gestation in a tertiary obstetric hospital in Perth, Western Australia. Milk samples, 24-hour milk production, and breast health data were collected every second day to day 10 postpartum, then every third day until infant discharge from the neonatal unit. Milk Na and K were measured at point of care (POC) using handheld ion selective meters, and Na:K calculated. Cultures were performed on postnatal days 8, 13, and every 6 days thereafter. For episodes of mastitis, milk was cultured at onset, and Na and Na:K measured daily until resolution. Women were followed up at 4 and 8 weeks postpartum. Results: In a sample of 44 women, 4 mastitis cases were detected in 3 women during their infants' neonatal stay; all had elevated milk Na and Na:K that resolved within 48 hours; 2/4 experienced reduced milk production and 1/4 had heavy growth of Staphylococcus epidermidis. A further 2 mastitis cases were reported in 39 women followed up to 8 weeks postpartum. Four women had elevated milk Na and Na:K without clinical signs of mastitis; three also had reduced milk production. Conclusions: POC testing of milk Na and/or Na:K may offer a useful indicator of breast health. Mastitis may cause an acute reduction in milk production regardless of the presence of culture-positive infection.
Collapse
Affiliation(s)
- Sharon Lisa Perrella
- School of Molecular Science, The University of Western Australia, Crawley, Western Australia, Australia
| | - Emma-Lee Anderton-May
- Neonatology Clinical Care Unit, King Edward Memorial Hospital, Subiaco, Western Australia, Australia
| | - Grace McLoughlin
- School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Ching Tat Lai
- School of Molecular Science, The University of Western Australia, Crawley, Western Australia, Australia
| | - Karen Norrie Simmer
- Neonatology Clinical Care Unit, King Edward Memorial Hospital, Subiaco, Western Australia, Australia.,School of Medicine, The University of Western Australia, Crawley, Western Australia, Australia
| | - Donna Tracy Geddes
- School of Molecular Science, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
15
|
The Dietary Inflammatory Index Is Associated with Subclinical Mastitis in Lactating European Women. Nutrients 2022; 14:nu14224719. [PMID: 36432405 PMCID: PMC9696022 DOI: 10.3390/nu14224719] [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] [Received: 09/23/2022] [Revised: 10/20/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Subclinical mastitis (SCM) is an inflammatory state of the lactating mammary gland, which is asymptomatic and may have negative consequences for child growth. The objectives of this study were to: (1) test the association between the dietary inflammatory index (DII®) and SCM and (2) assess the differences in nutrient intakes between women without SCM and those with SCM. One hundred and seventy-seven women with available data on human milk (HM) sodium potassium ratio (Na:K) and dietary intake data were included for analysis. Multivariable logistic regression was used to examine the association between nutrient intake and the DII score in relation to SCM. Women without SCM had a lower median DII score (0.60) than women with moderate (1.12) or severe (1.74) SCM (p < 0.01). A one-unit increase in DII was associated with about 41% increased odds of having SCM, adjusting for country and mode of delivery, p = 0.001. Women with SCM had lower mean intakes of several anti-inflammatory nutrients. We show for the first time exploratory evidence that SCM may be associated with a pro-inflammatory diet and women with SCM have lower intakes of several antioxidant and anti-inflammatory nutrients.
Collapse
|
16
|
Lactobacillus supports Clostridiales to restrict gut colonization by multidrug-resistant Enterobacteriaceae. Nat Commun 2022; 13:5617. [PMID: 36153315 PMCID: PMC9509339 DOI: 10.1038/s41467-022-33313-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/09/2022] [Indexed: 11/10/2022] Open
Abstract
Infections by multidrug-resistant Enterobacteriaceae (MRE) are life-threatening to patients. The intestinal microbiome protects against MRE colonization, but antibiotics cause collateral damage to commensals and open the way to colonization and subsequent infection. Despite the significance of this problem, the specific commensals and mechanisms that restrict MRE colonization remain largely unknown. Here, by performing a multi-omic prospective study of hospitalized patients combined with mice experiments, we find that Lactobacillus is key, though not sufficient, to restrict MRE gut colonization. Lactobacillus rhamnosus and murinus increase the levels of Clostridiales bacteria, which induces a hostile environment for MRE growth through increased butyrate levels and reduced nutrient sources. This mechanism of colonization resistance, an interaction between Lactobacillus spp. and Clostridiales involving cooperation between microbiota members, is conserved in mice and patients. These results stress the importance of exploiting microbiome interactions for developing effective probiotics that prevent infections in hospitalized patients. Multidrug-resistant Enterobacteriaceae (MRE) represent a major threat for patients’ health. Here, the authors describe how cooperation between gut commensal bacteria (Lactobacillus spp. And Clostridiales) restrict MRE colonization in mice and patients
Collapse
|
17
|
Polveiro RC, Vidigal PMP, de Oliveira Mendes TA, Yamatogi RS, da Silva LS, Fujikura JM, Da Costa MM, Moreira MAS. Distinguishing the milk microbiota of healthy goats and goats diagnosed with subclinical mastitis, clinical mastitis, and gangrenous mastitis. Front Microbiol 2022; 13:918706. [PMID: 36090116 PMCID: PMC9453028 DOI: 10.3389/fmicb.2022.918706] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/01/2022] [Indexed: 11/24/2022] Open
Abstract
Mastitis, mainly caused by bacterial intramammary infections, is the main problem in the breeding of dairy animals. The inflammations of the mammary gland is separated by types of mastitis, being subclinical, clinical, and the most severe, gangrenous mastitis. Here, we used 16S rRNA amplicon sequencing to characterize the bacterial microbiota of goat milk in the different types of goat mastitis caused by bacteria. We used 72 goat milk samples from a region of the state of Minas Gerais in Brazil, of which 12 were from clinically healthy animals, 42 from animals diagnosed with subclinical mastitis, 16 from animals with clinical mastitis, and 2 from animals with gangrenous mastitis. The group related to gangrenous mastitis was the most divergent in terms of alpha and beta diversity. The most abundant genus among samples of the groups was Staphylococcus spp., and we found a high abundance of Mycoplasma sp. in the milk of animals diagnosed with clinical mastitis. The most statistically relevant microorganisms among the groups were Prevotella sp., Ruminococcaceae, Prevotella ruminicola sp., and Providencia sp. We highlight a new association of bacterial agents in gangrenous mastitis among Escherichia sp./Shigella sp. and Enterococcus sp. and provide the second report of the genus Alkalibacterium sp., in milk samples. Only the taxa Staphylococcus sp., Bacteroides sp., Enterococcus, and Brevidabacterium sp., were present in all groups. The superpathway of L-tryptophan biosynthesis metabolites and the sucrose degradation III (sucrose invertase) pathway were the most prominent ones among the groups. In this study, we demonstrate how a rich microbiota of goat milk from healthy animals can be altered during the aggravation of different types of mastitis, in addition to demonstrating new bacterial genera in milk not previously detected in other studies as well as new associations between agents.
Collapse
Affiliation(s)
- Richard Costa Polveiro
- Laboratório de Doenças Bacterianas, Setor de Medicina Veterinária Preventiva e Saúde Pública, Departamento de Veterinária, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Pedro Marcus Pereira Vidigal
- Núcleo de Análise de Biomoléculas (NuBioMol), Centro de Ciências Biológicas, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | | - Ricardo Seiti Yamatogi
- Laboratório de Doenças Bacterianas, Setor de Medicina Veterinária Preventiva e Saúde Pública, Departamento de Veterinária, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | | | - Juliana Miwa Fujikura
- Laboratório de Doenças Bacterianas, Setor de Medicina Veterinária Preventiva e Saúde Pública, Departamento de Veterinária, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Mateus Matiuzzi Da Costa
- Departamento de Zootecnia, Universidade Federal do Vale do São Francisco, Petrolina, Pernambuco, Brazil
| | - Maria Aparecida Scatamburlo Moreira
- Laboratório de Doenças Bacterianas, Setor de Medicina Veterinária Preventiva e Saúde Pública, Departamento de Veterinária, Universidade Federal de Viçosa, Viçosa, MG, Brazil
- *Correspondence: Maria Aparecida Scatamburlo Moreira,
| |
Collapse
|
18
|
Hoque MN, Rahman MS, Islam T, Sultana M, Crandall KA, Hossain MA. Induction of mastitis by cow-to-mouse fecal and milk microbiota transplantation causes microbiome dysbiosis and genomic functional perturbation in mice. Anim Microbiome 2022; 4:43. [PMID: 35794639 PMCID: PMC9258091 DOI: 10.1186/s42523-022-00193-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 06/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mastitis pathogenesis involves a wide range of opportunistic and apparently resident microorganims including bacteria, viruses and archaea. In dairy animals, microbes reside in the host, interact with environment and evade the host immune system, providing a potential for host-tropism to favor mastitis pathogenesis. To understand the host-tropism phenomena of bovine-tropic mastitis microbiomes, we developed a cow-to-mouse mastitis model. METHODS A cow-to-mouse mastitis model was established by fecal microbiota transplantation (FMT) and milk microbiota transplantation (MMT) to pregnant mice to assess microbiome dysbiosis and genomic functional perturbations through shotgun whole metagenome sequencing (WMS) along with histopathological changes in mice mammary gland and colon tissues. RESULTS The cow-to-mouse FMT and MMT from clinical mastitis (CM) cows induced mastitis syndromes in mice as evidenced by histopathological changes in mammary gland and colon tissues. The WMS of 24 samples including six milk (CM = 3, healthy; H = 3), six fecal (CM = 4, H = 2) samples from cows, and six fecal (CM = 4, H = 2) and six mammary tissue (CM = 3, H = 3) samples from mice generating 517.14 million reads (average: 21.55 million reads/sample) mapped to 2191 bacterial, 94 viral and 54 archaeal genomes. The Kruskal-Wallis test revealed significant differences (p = 0.009) in diversity, composition, and relative abundances in microbiomes between CM- and H-metagenomes. These differences in microbiome composition were mostly represented by Pseudomonas aeruginosa, Lactobacillus crispatus, Klebsiella oxytoca, Enterococcus faecalis, Pantoea dispersa in CM-cows (feces and milk), and Muribaculum spp., Duncaniella spp., Muribaculum intestinale, Bifidobacterium animalis, Escherichia coli, Staphylococcus aureus, Massilia oculi, Ralstonia pickettii in CM-mice (feces and mammary tissues). Different species of Clostridia, Bacteroida, Actinobacteria, Flavobacteriia and Betaproteobacteria had a strong co-occurrence and positive correlation as the indicator species of murine mastitis. However, both CM cows and mice shared few mastitis-associated microbial taxa (1.14%) and functional pathways regardless of conservation of mastitis syndromes, indicating the higher discrepancy in mastitis-associated microbiomes among lactating mammals. CONCLUSIONS We successfully induced mastitis by FMT and MMT that resulted in microbiome dysbiosis and genomic functional perturbations in mice. This study induced mastitis in a mouse model through FMT and MMT, which might be useful for further studies- focused on pathogen(s) involved in mastitis, their cross-talk among themselves and the host.
Collapse
Affiliation(s)
- M Nazmul Hoque
- Department of Gynecology, Obstetrics and Reproductive Health, Bangabandhu Sheikh Mujibur Rahman Agricultural University (BSMRAU), Gazipur, 1706, Bangladesh
| | - M Shaminur Rahman
- Department of Microbiology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Tofazzal Islam
- Institute of Biotechnology and Genetic Engineering (IBGE), BSMRAU, Gazipur, 1706, Bangladesh
| | - Munawar Sultana
- Department of Microbiology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Keith A Crandall
- Computational Biology Institute and Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
| | - M Anwar Hossain
- Department of Microbiology, University of Dhaka, Dhaka, 1000, Bangladesh.
- Jashore University of Science and Technology, Jashore, 7408, Bangladesh.
| |
Collapse
|
19
|
Morales-Ferré C, Franch À, Castell M, Olivares M, Rodríguez-Lagunas MJ, Pérez-Cano FJ. Staphylococcus epidermidis' Overload During Suckling Impacts the Immune Development in Rats. Front Nutr 2022; 9:916690. [PMID: 35859758 PMCID: PMC9289531 DOI: 10.3389/fnut.2022.916690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/13/2022] [Indexed: 11/20/2022] Open
Abstract
Mastitis is an inflammation of the mammary gland occurring in 3-33% of the breastfeeding mothers. The majority of mastitis cases have an infectious etiology. More than 75% of infectious mastitis are caused by Staphylococcus epidermidis and Staphylococcus aureus and involves breast milk microbiota alteration, which, may have an impact in lactating infant. The aim of this study was to analyze in rats during the suckling period and later in life the impact of a high and a low overload of Staphylococcus epidermidis, similarly as it occurs during the clinical and the subclinical mastitis, respectively. From days 2 to 21 of life, suckling rats were daily supplemented with low (Ls group) or high (Hs group) dose of S. epidermidis. Body weight and fecal humidity were periodically recorded. On days 21 and 42 of life, morphometry, hematological variables, intestinal gene expression, immunoglobulin (Ig) and cytokine profile and spleen cells' phenotype were measured. Although no differences were found in body weight, Ls and Hs groups showed higher body length and lower fecal humidity. Both doses induced small changes in lymphocytes subpopulations, reduced the plasma levels of Ig and delayed the Th1/Th2 balance causing a bias toward the Th2 response. No changes were found in cytokine concentration. The low dose affected the Tc cells intestinal homing pattern whereas the high dose had an impact on the hematological variables causing leukocytosis and lymphocytosis and also influenced the intestinal barrier maturation. In conclusion, both interventions with Staphylococcus epidermidis overload during suckling, affects the immune system development in short and long term.
Collapse
Affiliation(s)
- Carla Morales-Ferré
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA-UB), Santa Coloma de Gramenet, Spain
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA-UB), Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA-UB), Santa Coloma de Gramenet, Spain
| | | | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA-UB), Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA-UB), Santa Coloma de Gramenet, Spain
| |
Collapse
|
20
|
Tang D, Cao F, Yan C, Fang K, Ma J, Gao L, Sun B, Wang G. Extracellular Vesicle/Macrophage Axis: Potential Targets for Inflammatory Disease Intervention. Front Immunol 2022; 13:705472. [PMID: 35769456 PMCID: PMC9234271 DOI: 10.3389/fimmu.2022.705472] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Extracellular vesicles (EVs) can regulate the polarization of macrophages in a variety of inflammatory diseases by mediating intercellular signal transduction and affecting the occurrence and development of diseases. After macrophages are regulated by EVs, they mainly show two phenotypes: the proinflammatory M1 type and the anti-inflammatory M2 type. A large number of studies have shown that in diseases such as mastitis, inflammatory bowel disease, Acute lung injury, and idiopathic pulmonary fibrosis, EVs promote the progression of the disease by inducing the M1-like polarization of macrophages. In diseases such as liver injury, asthma, and myocardial infarction, EVs can induce M2-like polarization of macrophages, inhibit the inflammatory response, and reduce the severity of the disease, thus indicating new pathways for treating inflammatory diseases. The EV/macrophage axis has become a potential target for inflammatory disease pathogenesis and comprehensive treatment. This article reviews the structure and function of the EV/macrophage axis and summarizes its biological functions in inflammatory diseases to provide insights for the diagnosis and treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Desheng Tang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Changsheng Yan
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kun Fang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiamin Ma
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Gao
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Gang Wang,
| |
Collapse
|
21
|
Liu J, Wang J, Xu W, Zeng L, Wang C, An Y, Tian H, Zeng B, Dong Q, Ji Y, Gao X, Du G, Liu J, Su J, Xie H, Xie L. Amyloid fibril formation by casein and fatty acid composition in breast milk of mastitis patients. J Food Biochem 2022; 46:e14183. [PMID: 35383958 DOI: 10.1111/jfbc.14183] [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: 12/19/2021] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/29/2022]
Abstract
Mastitis can cause changes in the nutrient composition of breast milk, which may be harmful to both newborns and lactating mothers. In this study we preliminarily evaluated amyloid fibrils formation by casein and fatty acids (FA), as well as their potential relation with each other in the breast milk of mastitis patients. Six healthy volunteers and six mastitis patients were recruited from the Maternal and Child Health Care Hospital in Changchun were enrolled. Amyloid fibril content was assessed by thioflavin T fluorescence analysis, transmission electron microscope, circular dichroism, and proton nuclear magnetic resonance. FA contents were measured by gas chromatography. Healthy breast milk contained no amyloid fibrils but inflammatory breast milk did. Several FAs (hendecanoic acid, myristolenic acid, pentadecenoic acid, eicosatrienoic acid) differed significantly between the two groups (p < .05). The concentrations of the eicosatrienoic acid and eleven carbonic acids in the inflammatory groups were lower than those in the healthy groups, but the myristolenic acid and pentadecenoic acid were the opposite trend. Early detection of amyloid fibrils should be performed in lactating mothers with mastitis. Changes in FAs may reflect the importance of abnormal metabolism in amyloid fibril formation. PRACTICAL APPLICATIONS: The work preliminarily clarified the relationship between inflammation, fibril content, and fatty acid (FA) composition in breast milk. Healthy milk contained no amyloid fibril formed by casein but the inflammatory milk did. FAs were also significantly different between the two groups. Thus, an early determination of amyloid fibrils in milk should be considered for lactating women with mastitis to avoid the further malignant development. Additionally, the changes in FAs may reflect the importance of abnormal metabolism and oxidative pathways in amyloid fibril formation in the breast. Therefore, this study provided foundations for further investigation on the association between inflammation, fibril content and FA composition in breast milk.
Collapse
Affiliation(s)
- Jihua Liu
- Department of Natural Product Chemistry, Pharmacy College, Jilin University, Changchun, China
| | - Jia Wang
- Department of Natural Product Chemistry, Pharmacy College, Jilin University, Changchun, China
| | - Wenhui Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Lei Zeng
- Bethune Institute of Epigenetic Medicine, The First Hospital, Jilin University, Changchun, China
| | - Chunyu Wang
- State Key Laboratory of Supramolecular, Structure and Materials, Jilin University, Changchun, China
| | - Yang An
- Department of Natural Product Chemistry, Pharmacy College, Jilin University, Changchun, China
| | - Huimin Tian
- School of Nursing, Jilin University, Changchun, China
| | - Baohua Zeng
- Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Qinghai Dong
- Department of Natural Product Chemistry, Pharmacy College, Jilin University, Changchun, China
| | - Ye Ji
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Xiaojun Gao
- Department of Nephrology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Guangguang Du
- Department of Natural Product Chemistry, Pharmacy College, Jilin University, Changchun, China
| | - Jiayin Liu
- Department of Natural Product Chemistry, Pharmacy College, Jilin University, Changchun, China
| | - Jun Su
- Department of Natural Product Chemistry, Pharmacy College, Jilin University, Changchun, China
| | - Hongliu Xie
- Department of Natural Product Chemistry, Pharmacy College, Jilin University, Changchun, China
| | - Lin Xie
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
22
|
The hidden universe of human milk microbiome: origin, composition, determinants, role, and future perspectives. Eur J Pediatr 2022; 181:1811-1820. [PMID: 35124754 PMCID: PMC9056486 DOI: 10.1007/s00431-022-04383-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/14/2022]
Abstract
UNLABELLED Although traditionally considered sterile, human milk is currently recognized as an alive ecosystem that harbors not only bacteria, but also viruses, fungi and yeasts, and minor genera, collectively known as the human milk microbiome (HMM). The seeding of HMM is a complex phenomenon whose dynamics are still a matter of research. Many factors contribute to its determination, both maternal, neonatal, environmental, and related to human milk itself. The transmission of microorganisms to the infant through breastfeeding may impact its present and future health, mainly shaping the GI tract microbiome and immune system. The existence and persistence of HMM as a conserved feature among different species may also have an evolutionary meaning, which will become apparent only in evolutionary times. CONCLUSION The complexities of HMM warrant further research in order to deepen our knowledge on its origin, determinants, and impact on infants' health. The practical and translational implications of research on HMM (e.g., reconstitution of donor human milk through inoculation of infant's own mother milk, modulation of HMM through maternal dietary supplementation) should not be overlooked. WHAT IS KNOWN • Human milk harbors a wide variety of microorganisms, ranging from bacteria to viruses, fungi and yeasts, and minor genera. • Human milk microbiome is shaped over time by many factors: maternal, neonatal, environmental, and related to human milk itself. • The transmission of microorganisms through breastfeeding may impact the infant's present and future health. WHAT IS NEW • We provide an overview on human milk microbiome, hopefully encouraging physicians to consider it among the other better-known breastfeeding benefits. • Further studies, with standardized and rigorous study designs to enhance accuracy and reproducibility of the results, are needed to deepen our knowledge of the human milk microbiota and its role in newborn and infant's health.
Collapse
|
23
|
Abstract
Aside from nutritional components, human milk is rich in microorganisms. Through breastfeeding these microorganisms are introduced to the infant gut where they may transiently or persistently colonize it. Therefore, the human milk microbiota may be an important factor which shapes the infant gut microbiota further influencing infant health and disease. In the current review we aim to give a brief updated insight into the putative origin of the human milk microbiota, its constituents and the possible factors that shape it. Understanding the factors that determine the human milk microbiota composition and function will aid developing optimal postnatal feeding and intervention strategies to reduce the risk of communicable and noncommunicable diseases.
Collapse
Affiliation(s)
- Anastasia Mantziari
- Functional Foods Forum, Faculty of Medicine, University of Turku, Itäinen Pitkäkatu 4A, 20520 Turku, Finland
| | - Samuli Rautava
- University of Helsinki and Helsinki University Hospital, New Children's Hospital, Pediatric Research Center, Helsinki, Finland.
| |
Collapse
|
24
|
Selma-Royo M, Calvo Lerma J, Cortés-Macías E, Collado MC. Human milk microbiome: From actual knowledge to future perspective. Semin Perinatol 2021; 45:151450. [PMID: 34274151 DOI: 10.1016/j.semperi.2021.151450] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human milk is the gold standard for infant nutrition during the first months of life since it is perfectly adapted to the neonatal nutritional requirements and supports infant growth and development. Human milk contains a complex nutritional and bioactive composition including microorganisms and oligosaccharides which would also contribute to the gut and immune system maturation. Despite the growing evidence, the factors contributing to milk microbes' variations and the potential functions on the infant's gut are still uncovered. This short-review provides a general overview of milk microbiota, potential factors shaping its composition, contribution to the infant microbiota and immune system development, including the suggested biological relevance for infant health as well as the description of tools and strategies aimed to restore and module microbes in milk.
Collapse
Affiliation(s)
- Marta Selma-Royo
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain).
| | - Joaquim Calvo Lerma
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain)
| | - Erika Cortés-Macías
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain)
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain).
| |
Collapse
|