Next Article in Journal
The Neurovascular Unit Dysfunction in Alzheimer’s Disease
Next Article in Special Issue
Fatty Acid Unsaturation Degree of Plasma Exosomes in Colorectal Cancer Patients: A Promising Biomarker
Previous Article in Journal
The Role of Tenascin C in Cardiac Reverse Remodeling Following Banding–Debanding of the Ascending Aorta
Previous Article in Special Issue
The Proteome and Citrullinome of Hippoglossus hippoglossus Extracellular Vesicles—Novel Insights into Roles of the Serum Secretome in Immune, Gene Regulatory and Metabolic Pathways
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exosomal Cargo May Hold the Key to Improving Reproductive Outcomes in Dairy Cows

1
Institute of Health and Biomedical Innovation—Centre for Children’s Health Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4029, Australia
2
Central Analytical Research Facility—Queensland University of Technology, Gardens Point, Brisbane, QLD 4000, Australia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(4), 2024; https://doi.org/10.3390/ijms22042024
Submission received: 28 January 2021 / Revised: 15 February 2021 / Accepted: 15 February 2021 / Published: 18 February 2021
(This article belongs to the Special Issue Extracellular Vesicles in Phylogeny)

Abstract

:
The reproductive status of dairy cows remains a challenge for dairy farmers worldwide, with impaired fertility linked to a significant reduction in herd profitability, due in part to impaired immunity, increased metabolic pressure, and longer postpartum anestrous interval (PPAI). Exosomes are nanovesicles released from a variety of cell types and end up in circulation, and carry proteins, bioactive peptides, lipids, and nucleic acids specific to the place of origin. As such, their role in health and disease has been investigated in humans and animals. This review discusses research into exosomes in the context of reproduction in dairy herds and introduces recent advances in mass-spectrometry (MS) based proteomics that have a potential to advance quantitative profiling of exosomal protein cargo in a search for early biomarkers of cattle fertility.

1. Introduction

Dairy cow fertility has been in decline for the past 20 years [1,2,3]. Selective breeding for milk production traits, negative energy balance (NEB), poor health or infection during the transition period (3 weeks before and the 3 weeks after calving), and early pregnancy loss have all been attributed to this decline [3,4,5]. These factors are thought to be linked but the underlying biological mechanisms responsible for these perturbations to reproductive performance have not yet been fully established.
Although it is widely accepted that increased metabolic pressure due to increased milk production is associated with poor reproductive outcomes, average producing cows may also experience reproductive challenges [6]. There are reports that discuss the lesser significance of increased milk production on fertility, and instead highlight genetic potential, nutritional intake, health status and farm management as major contributing factors to fertility status of the cow [7]. However, reliable predictors of future reproductive performance remain to be determined.Body condition scoring (BCS), and more recently BCS linked to timing of pubertal onset, is one of the few key indicators used by dairy farmers to manage and predict herd profitability [8].
Heifers can be separated into high- and low-fertility groups based on their genetic merit and other measurable physical traits [9]. However, this model has been found to be substandard when trying to address underlying causes of subfertility, and newer models expressing the extremes of the fertility spectrum have been developed in order to better explore the mechanisms responsible for the decline in calving rates over the past two decades. Although these newer models have allowed for improved sampling and study of the physiological stresses leading to poor reproductive performance, the biological mechanisms driving the disease process resulting in subfertility remain to be elucidated.
Exosomes, nanovesicles of ≈30–150 nm in diameter, can be isolated from the bodily fluids of dairy cows (e.g., blood plasma, milk, and follicular fluid), and present a unique opportunity to studying the molecular cues that underlie poor reproductive performance [10]. Exosomes are most commonly formed by the inward budding of multivesicular bodies (MVB) in the cell and begin as intraluminal vesicles (ILVs), and play a critical role in cell–cell signaling [11,12]. The molecular contents of circulating exosomes derived from the blood plasma and milk of dairy cows have been characterized to some extent, and contain, for example, proteins, mRNA, micro(mi)RNAs, and lipids [10,13]. It is possible that miRNA contained in the blood plasma exosomes of dairy cows serve as an epigenetic regulator of biological signaling pathways, including inflammation, which in turn may affect reproduction and development of the fetus during pregnancy [14]. Additionally, qualitative differences in proteomic exosomal cargo have been previously established in milk and plasma samples between high- and low-fertility dairy cows, and between cattle with and without uterine infection [15,16,17]. Quantitative differences in exosomal proteins between these high- and low-fertility groups are yet to be fully elucidated and may hold the key to identifying potential biomarkers for fertility. Exosomes contained in the blood plasma, for instance, can provide a systemic snapshot of valuable information about the health-status of the animal, which may be directly or indirectly related to reproductive status. This review will focus on the potential application of exosome-derived biomarkers to predict and lead to improved bovine reproduction in relation to key aspects of dairy cow fertility.

2. Exosomes

Formation and Function

Within the cell there is a complex protein synthesis and sorting pathway, whereby protein folding and glycosylation begin in the endoplasmic reticulum (ER). Mature and proproteins are further modified as they pass through the Golgi apparatus, and following this are transported via transport vesicles to early endosomes (see Figure 1, next page) [18]. Early endosomes mature further into late endosomes, whereby they are transported to the cell surface and exocytosed via direct fusion with the plasma membrane [19]. Endocytosed materials may also be transferred to late endosomes and transported to lysosomes, or recycled back to the cell surface [20]. Late endosomes contain nucleic acids, proteins, lipids, and trans-Golgi Network (TGN)-derived transport vesicles; hence they are also termed multivesicular bodies (MVBs) [21]. ILVs within MVBs are released as extracellular vesicles (EVs), a subpopulation of which are termed exosomes [18,22]. Proteins involved in MVB formation and cargo sorting (endosomal sorting complexes required for transport (ESCRT) pathway) and its accessory proteins are also typically found in exosomes [22,23]. Therefore, ESCRT proteins such as Tumor Suppressor Gene 101 (TSG101) are used experimentally as positive exosomal markers, as are members of the tetraspanin family (CD9, CD63, CD81); the latter of which have recently been implicated as important mediators in mammalian reproduction [22,24,25].
Exosomal molecular cargo can be endocytosed by target cells via a number of different mechanisms; direct receptor–ligand interaction, through cell surface adhesion molecules such as integrins or cadherins that initiate endocytosis, or by the opsonization of exosomes inducing phagocytosis in the recipient cell [26,27]. It has been suggested that the uptake of exosomes may also depend on the recipient-cell type, as a study involving exosomes isolated from various cancer cell lines demonstrated differences in uptake by recipient cells regardless of the cell type of exosomal origin [28]. This suggests that exosomes can interact with any cell type, independent of the cell from which they themselves are derived, albeit by different mechanisms of endocytosis. Interestingly, Sung et al. (2020) confirmed pathfinding behaviour of cells as they migrate towards exosomal tracks in 2D and 3D models, and created a double reporter system to follow the release, uptake, and acidification of exosomal deposits in internalized compartments containing exosomes [29]. The results of these studies present promising directions for future research when considering the use of exosomes for targeted therapeutics.
Whereas exosomes were historically thought to contain cellular waste, more recent exosomal profiling has resulted in the understanding that they are intrinsic to cell maintenance, cell–cell signaling, immune modulation, and progression of tumor-derived cells and metastasis [22]. This has led to research into their ability to carry biomarkers of disease in easily attainable biological fluids such as blood, saliva, and urine [30,31,32,33], and their potential as therapeutic targets and delivery vehicles [26,30,34]. Currently, researchers have begun to establish EV profiles that will assist in determining the proportions of the various EV subtypes in any given biological sample, with the aim to better understand heterogenous populations of EVs and their distinct functions [35,36].

3. Bovine Reproduction

The reproductive health of dairy cows has been associated with a number of physiological factors and environmental factors. Heat stress has been implicated as an epigenetic modifier than may negatively impact upon the reproductive status of offspring [38,39], while NEB has been linked to poor transition around the time of calving and metabolic stress [40,41]. Importantly, non-esterified fatty acid (NEFA) surplus as a result of NEB has been shown to result in poor immune function and increased likelihood of uterine infection [40]. Inflammatory mediators from the prostaglandin (PG) family are known to play a part in reproductive processes in cattle, and as such have been the subject of investigations surrounding impaired fertility in dairy herds [42]. Qin and colleagues (2020) examined the effects of high NEFA concentrations on PG production in bovine endometrial (BEND) cells and observed decreased levels of prostaglandin E2 (PGE2) and prostaglandin F (PGF) in cell culture media supernatant compared to controls [43]. Similarly, cows with metritis were found to have a differential abundance of common uterine bacteria compared with healthy cows [44]. Researchers have therefore attempted to establish ways to better manage cattle during times of physiological and metabolic challenge in hopes of improving reproductive health. For example, micronutrient supplementation during the transition period improved outcomes without altering the methylation state of the cows [45]. Thus, factors affecting reproductive performance of dairy herds are various and complex, and ways of determining intervention at an earlier stage may improve outcomes at a minimal cost to farmers and herds.
Exosomes have been the focus of bovine studies examining effects on implantation and embryo development. Two separate studies confirmed that exosomes derived from the bovine uterus increased gene and protein expression of the pregnancy-recognition-associated protein interferon-tau (IFN-τ) when cocultured with bovine embryos in vitro [46,47]. Another study implicated a role in exosome secretion from both conceptus and endometrium in facilitating crosstalk during the attachment period, while exosomes derived from follicular fluid have been shown to improve oocyte competence and resistance to environmental stressors such as heat shock [48,49]. Collectively, these studies suggest that exosomes are widely involved in bovine reproduction, thus supporting further evaluation of their contents and function.
While the protein cargo of exosomes has been somewhat characterized qualitatively, larger scale in-depth studies of quantitative differences between high- and low-fertility groups have not been conducted [13,17,50,51]. Dysregulation of the immune system, metabolic perturbations around the time of calving, and impaired embryonic-maternal crosstalk during implantation have all been associated with poor reproductive outcomes, and all of which exosomes are known to play a part [2,6,13,17,47,52]. Quantitative differences in exosomal protein cargo may have a significant impact on the overall health of dairy cows, upon which fertility may be directly or indirectly impacted. Differences may also serve as a valuable tool for predicting reproductive outcomes early on in the life of the cow and warrants further investigation.

3.1. The Immune System

Successful reproduction in dairy cows relies on a competent immune system, especially during the periparturient period. Compromised immunity is associated with poor transition during the calving period and significant physiological stress, resulting in increased risk of postpartum uterine infection, mastitis, and an extended postpartum anestrous interval (PPAI). Studies have focused on various aspects of the immune system to better understand reproductive failings around early embryonic loss, postpartum uterine infection, and associated poor reproductive outcomes. Exosomes carry lipid mediators derived from arachidonic acid (AA), and enzymes involved in their synthesis, including inflammatory mediators associated with reproduction [53,54,55]. For example, PGs are small lipid compounds classed as eicosanoids, which among a diverse number of actions can behave as inflammatory mediators that are not only upregulated during infection and inflammation, but also play a critical role in establishment and maintenance of pregnancy in cattle [42,56,57]. PGE2 and PGF are responsible for establishing or inhibiting bovine pregnancy, respectively [42]. Upregulation of inflammatory pathways during critical time points in the reproductive cycle of dairy cows could therefore have a severe impact on their reproductive health (see Figure 2). In an in vitro model of uterine inflammation, PGE2 and PGF were found to be differentially expressed by bovine endometrial epithelial (bEEL) and stromal (bCSC) cells when exposed to inflammatory stimuli [58]. In further experiments, bEEL expression of PGF was increased when coincubated with plasma exosomes derived from dairy cows with uterine infection [51]. Fatty acid cyclooxygenase-2 (COX2), which is upstream of the proinflammatory PGE2, has been highlighted as a potential target for therapies including the use of nonsteroidal anti-inflammatory drugs (NSAIDs) (see Figure 2) [2,40], although NSAIDS have previously been found to be ineffectual on Cox2 mRNA levels [59]. Interestingly, NSAIDS were successful in inhibiting lipopolysaccharide (LPS)-induced PGE2 and tumor necrosis factor-alpha (TNFα) mRNA production, indicating a mechanism of action separate to Cox2 activity [59]. A recent meta-analysis aimed to compare antibiotic with non-antibiotic methods (e.g., NSAIDs) of treatment for acute puerperal metritis (APM) in postpartum cattle [60]. Unfortunately, due to a shortage of comparable studies, the researchers were unable to perform the analysis for non-antibiotic methods, therefore the use of NSAIDs to treat postpartum uterine infection in cattle remains largely unverified.

3.2. The Transition Period

The transition period is a demanding phase in the life of dairy cows and challenging from the farm management perspective. It is typically defined as the period ranging from 3 weeks before and after calving [61] and represents a time of metabolic stress for the dairy cow, as the animal undergoes immense physiological changes in preparation for and during early lactation. Dairy cows that have been selectively bred for milk production traits experience greater metabolic pressure associated with increased milk production. Subsequently, this results in a greater incidence of postpartum uterine infection and mastitis, leading to ongoing health issues and negative implications for further reproduction [1,2,41,62]. Markers of metabolic distress such as β-hydroxybutyrate (BHB), triacylglycerols (TAG) and fatty acids (FA) were found to be altered in the blood plasma [2,8,61]. In addition to this, hypocalcemia resulting in ‘milk fever’ can occur, which results in the death of approximately 1 in 20 affected cows, reduces both the productive lifespan and milk production with each milk fever episode, and comes with associated costs of treatment and prevention [1,41,63]. The impact of metabolic distress during the transition period on future calving is of interest to reproductive studies. Increased metabolic pressure around the time of calving leads to lengthened PPAI and pre- and postovulatory dysfunction, which can significantly delay return to estrous and time to mating and is therefore of major concern to dairy farmers who operate under a seasonal-calving pasture-based system [2,8].
Numerous studies have focused on the link between BCS, NEB, and feed-intake during the transition period as a method of immunomodulation, in hopes of improving management of the transition dairy cow [64,65,66,67]. The use of exosomes as a potential source of biomarkers for low- versus high-risk populations of dairy cows has been investigated, with promising, although inconclusive, results [13]. Exosomes derived from the blood plasma of healthy versus dairy cows with cytological endometritis have been found to differ in protein composition when analyzed by liquid chromatography–mass spectrometry (LC-MS), which included proteins associated with innate immunity, acute immune response, and immune regulation [68]. Similarly, an in vitro study applied blood plasma exosomes isolated from dairy cows with and without uterine infection to endometrial cell lines to study their effects on PG production and found a decrease in luteolytic promoter PGF produced by cells treated with exosomes derived from the infected cows [51]. This suggests the involvement of PGF in disrupting normal reproductive processes and offers a potential target for improving outcomes in these animals. Despite this, the transition period still proves to be a challenging time for dairy farmers and their herds, and further research is required to better identify at-risk cows in hopes of preventing postpartum infection and maintaining reproductive efficiency.
Thus far, partly due to the ethical nature of conducting in vivo experiments, studies have steered towards in vitro modeling of bovine uterine infection. However, this may not be representative of the full spectrum of physiological mechanisms involved in, and leading to, high- or low-fertility and susceptibility to reproductive disruption in early life and during the transition or postpartum period. Bodily fluid samples obtained from cattle with and without disease may already be compromised regarding differences in molecular content, as it would be expected that inflammatory/disease markers would be present in affected animals at the time of disease occurrence. A more useful and predictive method of testing for differences would require sampling at the baseline stage, long before cattle experience reproductive and immune challenges. For example, sampling may occur around the time of puberty or earlier in order to establish a predictive model of reproductive performance and predisposition for disease in the early stages of reproductive life. Currently, Fertility Breeding Value (FBV) and BCS are the only tools available to dairy farmers to assist in the herd selection process, which does not consider the individual genetics or physiology of animals, but merely relies on physical attributes and genetic lineage as predictors [9,69]. Early biomarkers of fertility would aim to provide the dairy industry with reliable data that can assist in herd selection and lessen the burden of operational costs associated with poor reproductive performance. While lipid and inflammatory mediators transported by exosomes have been linked to reproduction in cattle, differences in protein cargo may give a better understanding of cattle fertility and the mechanisms that underlie perturbations to healthy reproduction.

4. Epigenetics of Reproduction

Epigenetic regulation of gene expression has been well studied with regards to mammalian development [70,71,72,73,74]. However, a new area of epigenetics is developing following research into the role of miRNAs as epigenetic modulators and has been reviewed recently [75,76]. Briefly, the epigenome is controlled at the base level by the expression of genes that encode for a group of enzymes, termed DNA methyltransferases (DNMTs) [71,73]. DNMTs catalyze the transfer of methyl groups to a specific part of DNA—CpG islands—as a way of altering gene expression [71]. miRNA performs modulatory actions at the epigenetic level by targeting DNMTs and histone deacetylases (HDACs) [75]. miRNA also has a direct impact on protein abundance via regulation at the translational level. Binding of miRNA to 3′ untranslated regions (UTRs) of target mRNA transcripts results in gene silencing or degradation, dependent on whether binding is imperfectly matched to the target sequence, or complimentary [77]. The epigenetic–miRNA regulatory loop also controls miRNA expression through DNA methylation, histone modification and RNA, and aberrations to these control mechanisms are associated with pathological health states [75,78,79]. Researchers have started to explore differential miRNA expression in hopes of finding early biomarkers of disease [80,81,82].
Bovine blood sera and exosomes have been subjected to miRNA profiling, and while 282 shared miRNAs were identified, 12 miRNAs were found to be differentially expressed between sera and exosomes [83]. Circulating miRNA has been shown to be a predictor of early pregnancy [84,85], and exosomal miRNA an indicator of early pregnancy loss in a cloned cattle study using somatic cell nuclear transfer (SCNT)-derived embryos [86]. The bovine estrous cycle, oocytes and preimplantation embryos have also been studied with regards to their specific exosomal and cell-free miRNA profiles. Subsequently, it was found that differential miRNA expression occurs during various stages of the estrous cycle and altered miRNA expression is associated with developmental competence of both oocytes and embryos [87,88,89,90]. Collectively, these results suggest that miRNA of exosomal and circulating origin may play an important role in regulating bovine reproduction. Correlative studies between miRNA and protein abundance would provide a comprehensive overview of the mechanisms behind systemic and local molecular regulation linked to reproductive outcomes.

5. Proteomics of Exosomes Derived from Bodily Fluids

5.1. Mass Spectrometry

Mass spectrometry (MS) is the technique of choice for determining the abundance of hundreds to thousands of proteins and continues to evolve through advancements in instrumentation, data acquisition modes and data analysis software. Its utility in protein analysis has a long history and has been extensively reviewed elsewhere [91,92,93,94]. In brief, methods for the effective formation of molecular ions from liquid or gas were established in the 1980s, and subsequently this led to the development of mass analyzers that were capable of determining the mass or structure of polypeptides with a high degree of sensitivity and accuracy [95,96,97,98]. MS systems are now commonly integrated and coupled with LC (LC-MS), which is the preferred method for analyzing samples with a high degree of complexity [98]. Initially widely used for peptide and protein identification in data-dependent acquisition (DDA) studies, MS instruments are now capable of peptide quantitation by labeled, relative, or targeted (absolute quantitation) methods, termed data-independent acquisition (DIA) [91,93,99,100].
In relation to dairy cow reproduction, MS has been utilized to perform thorough and reproducible analyses of bovine plasma, milk, follicular fluid, and uterine flushings [101,102,103]. To provide a better understanding of the signaling pathways associated with reproduction, exosomes isolated from milk of dairy cows have also been analyzed using a range of MS strategies in a number of studies [16,104,105,106]. Additionally, charge detection mass spectrometry (CDMS) and label-free spectral counting have been used successfully to characterize and quantify exosomes from milk and colostrum [104,105,107], and both milk and plasma exosomes have undergone qualitative analysis in DDA studies [4,16]. What is currently lacking in the field is a thorough quantitative analysis of the bovine blood plasma exosomal proteome, which may provide a better systemic snapshot of overall health and pathways associated with fertility, and thus clues to reproductive status in dairy cows. Table 1 summarizes what is currently known, and what remains to be established in relation to the role of exosomes of various origin and dairy cow reproduction.
DDA and targeted methods of MS, while effective, can be costly and/or only applicable to a limited number of samples. More recently, techniques such as sequential window acquisition of all theoretical mass spectra (SWATH-MS), termed next-generation proteomics, have emerged that allow the analysis of a greater number of samples with greater quantitative precision and impressive proteome coverage [116,117,118].

5.2. Next-Generation Proteomics

First described by Gillet et al. (2012), SWATH-MS is a variant of DIA that has already been applied to a large number of proteomic studies, including the analysis of exosomal protein cargo [116,119,120,121]. A major advantage of SWATH-MS approach is that quantitation is conducted using fragment ions, which are collected for all ionizable peptides in a sample, irrespective of their abundance. This is achieved using wide precursor isolation windows, which cover the expected mass range of all precursor ions. This effectively eliminates a bias in quantitation that other proteomics strategies have typically suffered from and permits a larger number of proteins across larger cohorts of samples to be analyzed with fewer missing values [117,118]. In its original implementation introduced by Gillet and colleagues, the highly complex nature of SWATH-MS data is dealt with using spectral libraries, however more recently, algorithms for library-free analysis have been developed [119,122,123].
Compared to other quantitative proteomics methods, the ease at which data is acquired is also a significant advantage, as once the precursor isolation scheme is set and method optimized for a particular sample type, analysis of different samples of the same type can be performed using the same method. A collaborative study looking at reproducibility and accuracy of SWATH-MS data detected and quantified >4000 proteins from Human embryonic kidney 293 (HEK293) cells in a 2-h run, and this was reproducible across multiple laboratories [124]. This allows proteomics studies to be performed on a much larger scale than originally feasible, with a high level of reproducibility and accuracy similar to that of targeted methods, but without the constraints of one-time data acquisition, as has been previously demonstrated [124,125]. The most promising feature of SWATH-MS in agriculture is that the data generated is ideal for retrospective quantitative analysis. SWATH-MS data may be exploited by remining them for new insights as genomic databases improve or as new compositional questions arise such as the ones derived from epigenetics analysis.

5.3. Current Challenges

Irrespective of MS approach employed, the analysis will largely depend on sample processing prior to MS. Highly abundant exosome proteins could compromise quantitation of low abundant cargo proteins of reproductive tissue origin and thus of biomarker potential. Enrichment of exosome populations of interest are therefore key to a successful outcome. The current methods of exosome purification involve sequential centrifugation, ultrafiltration, and size-exclusion chromatography, although there is currently no ‘gold-standard’ for exosome isolation [126,127,128,129,130]. These strategies, however, do not enrich for specific populations of exosomes that may be carrying the information specific to compromised fertility in cattle and further enrichment may be required [35]. This becomes even more critical when analyzing exosomes from bodily fluids and, in particular, from blood plasma, where the presence of several highly abundant plasma proteins such as albumin, globulins and fibrinogen may limit the overall number of exosome proteins detected in the study [131]. Furthermore, in the case of multistep enrichment, reproducibility of exosome preparation will have a significant impact on the ability of MS-based methods to reflect a true link between protein abundance and a biological phenomenon under study.

6. Conclusions

Suboptimal fertility in dairy cows has been attributed to acquired conditions such as poor uterine health, the adaptation to the transition period, and maternal-embryonic crosstalk in early pregnancy. Fertility status in dairy cows may also be determined at a much earlier timepoint due to factors stemming from genetic variants, which manifests in vivo as alterations to signaling pathways related to reproduction. Whether the fertility is a result of an acquired condition or inherited, the body responds in-kind by releasing exosomes that contain bioactive cargo that may provide a clue to cattle fertility [17,50,51]. Exosome research is a rapidly developing area of investigation for diagnostic and prognostic purposes. The qualitative and quantitative difference between exosomal cargo associated with different physiological conditions is determined using numerous ‘omics’ technologies and quantitative MS is at the forefront of this research. Specifically, a next-generation proteomics approach that relies on SWATH data acquisition to explore biomarkers of fertility on exosomes isolated and enriched from bovine blood plasma is currently being undertaken (unpublished data). Future research will aim to build on this concept through the study of miRNA on cellular function and signaling pathways related to fertility status of the animal, in hopes of developing targeted therapeutics to improve reproductive performance in cattle.

Author Contributions

Conceptualization, N.T. and M.D.M.; investigation, N.T.; writing—original draft preparation, N.T.; writing—review and editing, P.S., P.A. and M.D.M.; visualization, N.T.; supervision, P.S. and M.D.M. All authors have read and agreed to the published version of the manuscript.

Funding

NT is supported by a student scholarship from the Australian Research Council (Grant No: ARC LP160101854) and QUT HDR Tuition fee sponsorship. Our laboratory experiments were funded, in part, by funding from a partnership fund (DRCX1302) between the New Zealand Ministry of Business, Innovation and Employment and New Zealand dairy farmers through DairyNZ Inc.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AAArachidonic acid
APMAcute puerperal metritis
BCSBody condition scoring
bCSCBovine stromal cells
bEELBovine endometrial epithelial cells
BHBβ-hydroxybutyrate
CDMSCharge detection mass spectrometry
COX1/Cox1Fatty acid cyclooxygenase-1
COX2/Cox2Fatty acid cyclooxygenase-2
NEBNegative energy balance
DDAData-dependent acquisition
DIAData-independent acquisition
DNMTDNA methyltransferase
EREndoplasmic reticulum
ESCRTEndosomal sorting complexes required for transport
EVExtracellular vesicles
FAFatty acids
FBVFertility breeding value
HDAChistone deacetylase
HEK293Human embryonic kidney 293
ILVIntraluminal vesicle
IFN-τInterferon-tau
LCLiquid chromatography
LPSLipopolysaccharide
miRNAmicroRNA
MSMass spectrometry
MVBMultivesicular bodies
NEBNegative energy balance
NEFANonesterified fatty acid
NSAIDNonsteroidal anti-inflammatory drug
PGProstaglandin
PGE2Prostaglandin E2
PGFProstaglandin F
PPAIPostpartum anestrous interval
SCNTSomatic cell nuclear transfer
SWATH-MSSequential window acquisition of all theoretical mass spectra
TAGTriacylglycerols
TGNtrans-Golgi network
TNFαTumor necrosis factor-alpha
TSG101Tumor Suppressor Gene 101
UTRUntranslated region

References

  1. Mitchell, M.D.; Crookenden, M.A.; Vaswani, K.; Roche, J.R.; Peiris, H.N. The frontiers of biomedical science and its application to animal science in addressing the major challenges facing Australasian dairy farming. Anim. Prod. Sci. 2020, 60. [Google Scholar] [CrossRef] [Green Version]
  2. Roche, J.R.; Burke, C.R.; Crookenden, M.A.; Heiser, A.; Loor, J.L.; Meier, S.; Mitchell, M.D.; Phyn, C.V.C.; Turner, S.-A. Fertility and the transition dairy cow. Reprod. Fertil. Dev. 2018, 30, 85. [Google Scholar] [CrossRef]
  3. Berry, D.P.; Friggens, N.C.; Lucy, M.C.; Roche, J.R. Milk Production and Fertility in Cattle. Annu. Rev. Anim. Biosci. 2016, 4, 269–290. [Google Scholar] [CrossRef] [PubMed]
  4. Koh, Y.Q.; Peiris, H.N.; Vaswani, K.; Almughlliq, F.B.; Meier, S.; Burke, C.R.; Roche, J.R.; Reed, C.B.; Arachchige, B.J.; Reed, S.; et al. Proteome profiling of exosomes derived from plasma of heifers with divergent genetic merit for fertility. J. Dairy Sci. 2018, 101, 6462–6473. [Google Scholar] [CrossRef]
  5. Garnsworthy, P.C.; Sinclair, K.D.; Webb, R. Integration of Physiological Mechanisms That Influence Fertility in Dairy Cows. Animal 2008, 2, 1144–1152. [Google Scholar] [CrossRef] [Green Version]
  6. Formigoni, A.; Trevisi, E. Transition Cow: Interaction with Fertility. Vet. Res. Commun. 2003, 27, 143–152. [Google Scholar] [CrossRef] [PubMed]
  7. Lucy, M.C. Reproductive Loss in High-Producing Dairy Cattle: Where Will It End? J. Dairy Sci. 2001, 84, 1277–1293. [Google Scholar] [CrossRef]
  8. Roche, J.F.; Mackey, D.; Diskin, M.D. Reproductive management of postpartum cows. Anim. Reprod. Sci. 2000, 60–61, 703–712. [Google Scholar] [CrossRef]
  9. Bowley, F.; Green, R.; Amer, P.; Meier, S. Novel approaches to genetic analysis of fertility traits in New Zealand dairy cattle. J. Dairy Sci. 2015, 98, 2005–2012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Mitchell, M.; Scholz-Romero, K.; Reed, S.; Peiris, H.; Koh, Y.; Meier, S.; Walker, C.; Burke, C.; Roche, J.; Rice, G.; et al. Plasma exosome profiles from dairy cows with divergent fertility phenotypes. J. Dairy Sci. 2016, 99, 7590–7601. [Google Scholar] [CrossRef] [Green Version]
  11. Raimondo, F.; Morosi, L.; Chinello, C.; Magni, F.; Pitto, M. Advances in membranous vesicle and exosome proteomics improving biological understanding and biomarker discovery. Proteomics 2011, 11, 709–720. [Google Scholar] [CrossRef]
  12. Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding Light on the Cell Biology of Extracellular Vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  13. Crookenden, M.; Walker, C.; Peiris, H.; Koh, Y.; Heiser, A.; Loor, J.; Moyes, K.; Murray, A.; Dukkipati, V.; Kay, J.; et al. Short communication: Proteins from circulating exosomes represent metabolic state in transition dairy cows. J. Dairy Sci. 2016, 99, 7661–7668. [Google Scholar] [CrossRef] [Green Version]
  14. Sohel, M.M.; Hoelker, M.; Noferesti, S.S.; Salilew-Wondim, D.; Tholen, E.; Looft, C.; Rings, F.; Uddin, M.J.; Spencer, T.E.; Schellander, K.; et al. Exosomal and Non-Exosomal Transport of Extra-Cellular Micrornas in Follicular Fluid: Implications for Bovine Oocyte Developmental Competence. PLoS ONE 2013, 8, e78505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Crookenden, M.A.; Walker, C.G.; Peiris, H.; Koh, Y.; Almughlliq, F.; Vaswani, K.; Reed, S.; Heiser, A.; Loor, J.J.; Kay, J.K.; et al. Effect of Circulating Exosomes from Transition Cows on Madin-Darby Bovine Kidney Cell Function. J. Dairy Sci. 2017, 100, 5687–5700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Koh, Y.Q.; Peiris, H.N.; Vaswani, K.; Meier, S.; Burke, C.R.; Macdonald, K.A.; Roche, J.R.; Almughlliq, F.; Arachchige, B.J.; Reed, S.; et al. Characterization of Exosomes from Body Fluids of Dairy Cows. J. Anim. Sci. 2017, 95, 3893–3904. [Google Scholar] [CrossRef] [PubMed]
  17. Almughlliq, F.B.; Koh, Y.Q.; Peiris, H.N.; Vaswani, K.; McDougall, S.; Graham, E.M.; Burke, C.R.; Arachchige, B.J.; Reed, S.; Mitchell, M.D. Proteomic content of circulating exosomes in dairy cows with or without uterine infection. Theriogenology 2018, 114, 173–179. [Google Scholar] [CrossRef] [PubMed]
  18. Hessvik, N.P.; Llorente, A. Current knowledge on exosome biogenesis and release. Cell. Mol. Life Sci. 2018, 75, 193–208. [Google Scholar] [CrossRef] [Green Version]
  19. Luzio, J.P.; Pryor, P.R.; Bright, N.A. Lysosomes: Fusion and function. Nat. Rev. Mol. Cell Biol. 2007, 8, 622–632. [Google Scholar] [CrossRef]
  20. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef]
  21. Stoorvogel, W.; Kleijmeer, M.J.; Geuze, H.J.; Raposo, G. The Biogenesis and Functions of Exosomes. Traffic 2002, 3, 321–330. [Google Scholar] [CrossRef]
  22. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Simons, M.; Raposo, G. Exosomes—Vesicular Carriers for Intercellular Communication. Curr. Opin. Cell Biol. 2009, 21, 575–581. [Google Scholar] [CrossRef] [PubMed]
  24. Jankovičová, J.; Neuerová, Z.; Sečová, P.; Bartóková, M.; Bubeníčková, F.; Komrsková, K.; Postlerová, P.; Antalíková, J. Tetraspanins in Mammalian Reproduction: Spermatozoa, Oocytes and Embryos. Med. Microbiol. Immunol. 2020, 209, 407–425. [Google Scholar] [CrossRef] [PubMed]
  25. Henne, W.M.; Buchkovich, N.J.; Emr, S.D. The ESCRT Pathway. Dev. Cell 2011, 21, 77–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Farooqi, A.A.; Desai, N.N.; Qureshi, M.Z.; Librelotto, D.R.N.; Gasparri, M.L.; Bishayee, A.; Nabavi, S.M.; Curti, V.; Daglia, M. Exosome biogenesis, bioactivities and functions as new delivery systems of natural compounds. Biotechnol. Adv. 2018, 36, 328–334. [Google Scholar] [CrossRef] [PubMed]
  27. Whiteside, T.L. Exosomes and tumor-mediated immune suppression. J. Clin. Investig. 2016, 126, 1216–1223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Horibe, S.; Tanahashi, T.; Kawauchi, S.; Murakami, Y.; Rikitake, Y. Mechanism of recipient cell-dependent differences in exosome uptake. BMC Cancer 2018, 18, 1–9. [Google Scholar] [CrossRef] [Green Version]
  29. Sung, B.H.; Von Lersner, A.; Guerrero, J.; Krystofiak, E.S.; Inman, D.; Pelletier, R.; Zijlstra, A.; Ponik, S.M.; Weaver, A.M. A live cell reporter of exosome secretion and uptake reveals pathfinding behavior of migrating cells. Nat. Commun. 2020, 11, 1–15. [Google Scholar]
  30. Rashed, M.H.; Bayraktar, E.; Helal, G.K.; Abd-Ellah, M.F.; Amero, P.; Chavez-Reyes, A.; Rodriguez-Aguayo, C. Exosomes: From Garbage Bins to Promising Therapeutic Targets. Int. J. Mol. Sci. 2017, 18, 538. [Google Scholar] [CrossRef] [Green Version]
  31. Han, Y.; Jia, L.; Zheng, Y.; Li, W. Salivary Exosomes: Emerging Roles in Systemic Disease. Int. J. Biol. Sci. 2018, 14, 633–643. [Google Scholar] [CrossRef]
  32. Meng, X.; Pan, J.; Sun, S.; Gong, Z. Circulating exosomes and their cargos in blood as novel biomarkers for cancer. Transl. Cancer Res. 2018, 7, S226–S242. [Google Scholar] [CrossRef]
  33. Panfoli, I. Cancer exosomes in urine: A promising biomarker source. Transl. Cancer Res. 2017, 6, S1389–S1393. [Google Scholar] [CrossRef]
  34. Nazimek, K.; Bryniarski, K.; Santocki, M.; Ptak, W. Exosomes as mediators of intercellular communication: Clinical implications. Pol. Arch. Intern. Med. 2015, 125, 370–380. [Google Scholar] [CrossRef]
  35. Kowal, J.; Arras, G.; Colombo, M.; Jouve, M.; Morath, J.P.; Primdal-Bengtson, B.; Dingli, F.; Loew, D.; Tkach, M.; Théry, C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl. Acad. Sci. USA 2016, 113, E968–E977. [Google Scholar] [CrossRef] [Green Version]
  36. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef]
  37. Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, Rna Cargo Selection, Content, Release, and Uptake; Springer: New York, NY, USA, 2016; pp. 301–312. [Google Scholar]
  38. Huber, E.; Notaro, U.; Recce, S.; Rodríguez, F.; Ortega, H.; Salvetti, N.; Rey, F. Fetal programming in dairy cows: Effect of heat stress on progeny fertility and associations with the hypothalamic-pituitary-adrenal axis functions. Anim. Reprod. Sci. 2020, 216, 106348. [Google Scholar] [CrossRef]
  39. Lee, J.; Lee, S.; Son, J.; Lim, H.; Kim, E.; Kim, D.; Ha, S.; Hur, T.; Lee, S.; Choi, I. Analysis of Circulating-Microrna Expression in Lactating Holstein Cows under Summer Heat Stress. PLoS ONE 2020, 15, e0231125. [Google Scholar] [CrossRef] [PubMed]
  40. Bradford, B.; Yuan, K.; Farney, J.; Mamedova, L.; Carpenter, A. Invited review: Inflammation during the transition to lactation: New adventures with an old flame. J. Dairy Sci. 2015, 98, 6631–6650. [Google Scholar] [CrossRef]
  41. Esposito, G.; Irons, P.C.; Webb, E.C.; Chapwanya, A. Interactions between negative energy balance, metabolic diseases, uterine health and immune response in transition dairy cows. Anim. Reprod. Sci. 2014, 144, 60–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Arosh, J.A.; Banu, S.K.; Kimmins, S.; Chapdelaine, P.; MacLaren, L.A.; Fortier, M.A. Effect of Interferon-Tau on Prostaglandin Biosynthesis, Transport, and Signaling at the Time of Maternal Recognition of Pregnancy in Cattle: Evidence of Polycrine Actions of Prostaglandin E2. Endocrinology 2004, 145, 5280–5293. [Google Scholar] [CrossRef] [Green Version]
  43. Qin, X.; Yang, S.; Zhang, Y.; Li, L.; Li, P.; Long, M.; Guo, Y. Effects of non-esterified fatty acids on relative abundance of prostaglandin E2 and F2α synthesis-related mRNA transcripts and protein in endometrial cells of cattle in vitro. Anim. Reprod. Sci. 2020, 221, 106549. [Google Scholar] [CrossRef] [PubMed]
  44. Chen, H.; Fu, K.; Pang, B.; Wang, J.; Li, H.; Jiang, Z.; Feng, Y.; Tian, W.; Cao, R. Determination of uterine bacterial community in postpartum dairy cows with metritis based on 16S rDNA sequencing. Vet. Anim. Sci. 2020, 10, 100102. [Google Scholar] [CrossRef] [PubMed]
  45. Gasselin, M.; Boutinaud, M.; Prézelin, A.; Debournoux, P.; Fargetton, M.; Mariani, E.; Zawadzki, J.; Kiefer, H.; Jammes, H. Effects of micronutrient supplementation on performance and epigenetic status in dairy cows. Animal 2020, 14, 2326–2335. [Google Scholar] [CrossRef]
  46. Qiao, F.; Ge, H.; Ma, X.; Zhang, Y.; Zuo, Z.; Wang, M.; Wang, Y. Bovine uterus-derived exosomes improve developmental competence of somatic cell nuclear transfer embryos. Theriogenology 2018, 114, 199–205. [Google Scholar] [CrossRef] [PubMed]
  47. Kusama, K.; Nakamura, K.; Bai, R.; Nagaoka, K.; Sakurai, T.; Imakawa, K. Intrauterine Exosomes Are Required for Bovine Conceptus Implantation. Biochem. Biophys. Res. Commun. 2018, 495, 1370–1375. [Google Scholar] [CrossRef]
  48. Rodrigues, T.A.; Tuna, K.M.; Alli, A.A.; Tribulo, P.; Hansen, P.J.; Koh, J.; Paula-Lopes, F.F. Follicular fluid exosomes act on the bovine oocyte to improve oocyte competence to support development and survival to heat shock. Reprod. Fertil. Dev. 2019, 31, 888. [Google Scholar] [CrossRef]
  49. Nakamura, K.; Kusama, K.; Bai, R.; Sakurai, T.; Isuzugawa, K.; Godkin, J.D.; Suda, Y.; Imakawa, K. Induction of IFNT-Stimulated Genes by Conceptus-Derived Exosomes during the Attachment Period. PLoS ONE 2016, 11, e0158278. [Google Scholar] [CrossRef] [Green Version]
  50. Almughlliq, F.B.; Koh, Y.Q.; Peiris, H.N.; Vaswani, K.; Holland, O.; Meier, S.; Roche, J.R.; Burke, C.R.; Crookenden, M.A.; Arachchige, B.J.; et al. Circulating Exosomes May Identify Biomarkers for Cows at Risk for Metabolic Dysfunction. Sci. Rep. 2019, 9, 1–12. [Google Scholar] [CrossRef] [Green Version]
  51. Almughlliq, F.B.; Koh, Y.Q.; Peiris, H.N.; Vaswani, K.; McDougall, S.; Graham, E.M.; Burke, C.R.; Mitchell, M.D. Effect of exosomes from plasma of dairy cows with or without an infected uterus on prostaglandin production by endometrial cell lines. J. Dairy Sci. 2017, 100, 9143–9152. [Google Scholar] [CrossRef] [Green Version]
  52. Saeed-Zidane, M.; Linden, L.; Salilew-Wondim, D.; Held, E.; Neuhoff, C.; Tholen, E.; Hoelker, M.; Schellander, K.; Tesfaye, D. Cellular and exosome mediated molecular defense mechanism in bovine granulosa cells exposed to oxidative stress. PLoS ONE 2017, 12, e0187569. [Google Scholar] [CrossRef] [Green Version]
  53. Giller, K.; Drews, B.; Bérard, J.; Kienberger, H.; Schmicke, M.; Frank, J.; Spanier, B.; Daniel, H.; Geisslinger, G.; Ulbrich, S.E. Bovine embryo elongation is altered due to maternal fatty acid supplementation. Biol. Reprod. 2018, 99, 600–610. [Google Scholar] [CrossRef]
  54. Zhang, N.; Wang, L.; Luo, G.; Tang, X.; Ma, L.; Zheng, Y.; Liu, S.; Price, C.A.; Jiang, Z. Arachidonic Acid Regulation of Intracellular Signaling Pathways and Target Gene Expression in Bovine Ovarian Granulosa Cells. Animals 2019, 9, 374. [Google Scholar] [CrossRef] [Green Version]
  55. Subra, C.; Grand, D.; Laulagnier, K.; Stella, A.; Lambeau, G.; Paillasse, M.; de Medina, P.; Monsarrat, B.; Perret, B.; Silvente-Poirot, S.; et al. Exosomes Account for Vesicle-Mediated Transcellular Transport of Activatable Phospholipases and Prostaglandins. J. Lipid Res. 2010, 51, 2105–2120. [Google Scholar] [CrossRef] [Green Version]
  56. Banu, S.K.; Arosh, J.A.; Chapdelaine, P.; Fortier, M.A. Expression of Prostaglandin Transporter in the Bovine Uterus and Fetal Membranes During Pregnancy1. Biol. Reprod. 2005, 73, 230–236. [Google Scholar] [CrossRef]
  57. Ledgard, A.M.; Meier, S.; Peterson, A.J. Evaluation of the uterine environment early in pregnancy establishment to characterise cows with a potentially superior ability to support conceptus survival. Reprod. Fertil. Dev. 2011, 23, 737–747. [Google Scholar] [CrossRef] [Green Version]
  58. Almughlliq, F.B.; Koh, Y.Q.; Peiris, H.N.; Vaswani, K.; Arachchige, B.J.; Reed, S.; Mitchell, M.D. Eicosanoid Pathway Expression in Bovine Endometrial Epithelial and Stromal Cells in Response to Lipopolysaccharide, Interleukin 1 Beta, and Tumor Necrosis Factor Alpha. Reprod. Biol. 2018, 18, 390–396. [Google Scholar] [CrossRef]
  59. Myers, M.J.; Scott, M.L.; Deaver, C.M.; Farrell, D.E.; Yancy, H.F. Biomarkers of inflammation in cattle determining the effectiveness of anti-inflammatory drugs. J. Vet. Pharmacol. Ther. 2010, 33, 1–8. [Google Scholar] [CrossRef]
  60. Haimerl, P.; Arlt, S.; Borchardt, S.; Heuwieser, W. Antibiotic treatment of metritis in dairy cows—A meta-analysis. J. Dairy Sci. 2017, 100, 3783–3795. [Google Scholar] [CrossRef] [Green Version]
  61. Drackley, J.K. Biology of Dairy Cows During the Transition Period: The Final Frontier? J. Dairy Sci. 1999, 82, 2259–2273. [Google Scholar] [CrossRef]
  62. Trevisi, E.; Minuti, A. Assessment of the innate immune response in the periparturient cow. Res. Vet. Sci. 2018, 116, 47–54. [Google Scholar] [CrossRef] [PubMed]
  63. Champness, D. Milk Fever (Hypocalcaemia) in Cows. 2007. Available online: https://www.lowlinecattleassoc.com.au/wp-content/uploads/pdf/Milk-Fever-%E2%80%93-Treatment-Prevention.pdf (accessed on 8 February 2021).
  64. Vailati-Riboni, M.; Kanwal, M.; Bulgari, O.; Meier, S.; Priest, N.; Burke, C.; Kay, J.; McDougall, S.; Mitchell, M.; Walker, C.; et al. Body condition score and plane of nutrition prepartum affect adipose tissue transcriptome regulators of metabolism and inflammation in grazing dairy cows during the transition period. J. Dairy Sci. 2016, 99, 758–770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Vailati-Riboni, M.; Farina, G.; Batistel, F.; Heiser, A.; Mitchell, M.; Crookenden, M.; Walker, C.; Kay, J.; Meier, S.; Roche, J.; et al. Far-off and close-up dry matter intake modulate indicators of immunometabolic adaptations to lactation in subcutaneous adipose tissue of pasture-based transition dairy cows. J. Dairy Sci. 2017, 100, 2334–2350. [Google Scholar] [CrossRef] [PubMed]
  66. Roche, J.R.; Heiser, A.; Mitchell, M.D.; Crookenden, M.A.; Walker, C.G.; Kay, J.K.; Riboni, M.V.; Loor, J.J.; Meier, S. Strategies to Gain Body Condition Score in Pasture-Based Dairy Cows During Late Lactation and the Far-Off Nonlactating Period and Their Interaction with Close-up Dry Matter Intake. J. Dairy Sci. 2017, 100, 1720–1738. [Google Scholar] [CrossRef]
  67. Crookenden, M.; Walker, C.; Heiser, A.; Murray, A.; Dukkipati, V.; Kay, J.; Meier, S.; Moyes, K.; Mitchell, M.; Loor, J.; et al. Effects of precalving body condition and prepartum feeding level on gene expression in circulating neutrophils. J. Dairy Sci. 2017, 100, 2310–2322. [Google Scholar] [CrossRef] [Green Version]
  68. Miller, B.A.; Brewer, A.; Nanni, P.; Lim, J.J.; Callanan, J.J.; Grossmann, J.; Kunz, L.; De Almeida, A.M.; Meade, K.G.; Chapwanya, A. Characterization of circulating plasma proteins in dairy cows with cytological endometritis. J. Proteom. 2019, 205, 103421. [Google Scholar] [CrossRef]
  69. Meier, S.; Fisher, B.; Eketone, K.; McNaughton, L.R.; Amer, P.R.; Beatson, P.; Bryant, J.R.; Dodds, K.G.; Spelman, R.; Roche, J.R.; et al. Calf and Heifer Development and the Onset of Puberty in Dairy Cows with Divergent Genetic Merit for Fertility. N. Z. Soc. Anim. Prod. Proc. 2017, 77, 205–210. [Google Scholar]
  70. Holliday, R. Epigenetics: A Historical Overview. Epigenetics 2006, 1, 76–80. [Google Scholar] [CrossRef] [Green Version]
  71. Reik, W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nat. Cell Biol. 2007, 447, 7. [Google Scholar] [CrossRef]
  72. Skinner, M.K. Role of epigenetics in developmental biology and transgenerational inheritance. Birth Defects Res. Part C Embryo Today Rev. 2011, 93, 51–55. [Google Scholar] [CrossRef] [Green Version]
  73. Smith, Z.D.; Meissner, A. DNA methylation: Roles in mammalian development. Nat. Rev. Genet. 2013, 14, 204–220. [Google Scholar] [CrossRef] [PubMed]
  74. Kiefer, J.C. Epigenetics in development. Dev. Dyn. 2007, 236, 1144–1156. [Google Scholar] [CrossRef] [PubMed]
  75. Yao, Q.; Chen, Y.; Zhou, X. The Roles of Micrornas in Epigenetic Regulation. Curr. Opin. Chem. Biol. 2019, 51, 11–17. [Google Scholar] [CrossRef] [PubMed]
  76. Abeysinghe, P.; Turner, N.; Garcia, I.M.; Mosaad, E.; Peiris, H.N.; Mitchell, M.D. The Role of Exosomal Epigenetic Modifiers in Cell Communication and Fertility of Dairy Cows. Int. J. Mol. Sci. 2020, 21, 9106. [Google Scholar] [CrossRef]
  77. He, L.; Hannon, G.J. MicroRNAs: Small RNAs with a big role in gene regulation. Nat. Rev. Genet. 2004, 5, 522–531. [Google Scholar] [CrossRef]
  78. Costa-Pinheiro, P.; Montezuma, D.; Henrique, R.; Jerónimo, C. Diagnostic and prognostic epigenetic biomarkers in cancer. Epigenomics 2015, 7, 1003–1015. [Google Scholar] [CrossRef]
  79. Dawson, M.A.; Kouzarides, T. Cancer Epigenetics: From Mechanism to Therapy. Cell 2012, 150, 12–27. [Google Scholar] [CrossRef] [Green Version]
  80. Humphries, B.; Wang, Z.; Yang, C. MicroRNA Regulation of Epigenetic Modifiers in Breast Cancer. Cancers 2019, 11, 897. [Google Scholar] [CrossRef] [Green Version]
  81. Huang, W.; Yan, Y.; Liu, Y.; Lin, M.; Ma, J.; Zhang, W.; Dai, J.; Li, J.; Guo, Q.; Chen, H.; et al. Exosomes with low miR-34c-3p expression promote invasion and migration of non-small cell lung cancer by upregulating integrin α2β1. Signal Transduct. Target. Ther. 2020, 5, 1–13. [Google Scholar] [CrossRef]
  82. Ediriweera, M.K.; Cho, S.K. Targeting Mirnas by Histone Deacetylase Inhibitors (Hdaci): Rationalizing Epigenetics-Based Therapies for Breast Cancer. Pharmacol. Ther. 2020, 206, 107437. [Google Scholar] [CrossRef]
  83. Zhao, K.; Liang, G.; Sun, X.; Guan, L.L. Comparative miRNAome analysis revealed different miRNA expression profiles in bovine sera and exosomes. BMC Genom. 2016, 17, 630. [Google Scholar] [CrossRef] [PubMed]
  84. Ioannidis, J.; Donadeu, F.X. Circulating miRNA signatures of early pregnancy in cattle. BMC Genom. 2016, 17, 184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Gebremedhn, S.; Salilew-Wondim, D.; Hoelker, M.; Held-Hoelker, E.; Neuhoff, C.; Tholen, E.; Schellander, K.; Tesfaye, D. Exploring maternal serum microRNAs during early pregnancy in cattle. Theriogenology 2018, 121, 196–203. [Google Scholar] [CrossRef] [PubMed]
  86. De Bem, T.H.C.; da Silveira, J.C.; Sampaio, R.V.; Sangalli, J.R.; Oliveira, M.L.F.; Ferreira, R.M.; Silva, L.A.; Perecin, F.; King, W.A.; Meirelles, F.V.; et al. Low Levels of Exosomal-Mirnas in Maternal Blood Are Associated with Early Pregnancy Loss in Cloned Cattle. Sci. Rep. 2017, 7, 1–11. [Google Scholar] [CrossRef] [PubMed]
  87. Ioannidis, J.; Donadeu, F.X. Circulating microRNA Profiles during the Bovine Oestrous Cycle. PLoS ONE 2016, 11, e0158160. [Google Scholar] [CrossRef] [Green Version]
  88. Pasquariello, R.; Manzoni, E.; Fiandanese, N.; Viglino, A.; Pocar, P.; Brevini, T.; Williams, J.; Gandolfi, F. Implications of miRNA expression pattern in bovine oocytes and follicular fluids for developmental competence. Theriogenology 2020, 145, 77–85. [Google Scholar] [CrossRef]
  89. Lin, X.; Beckers, E.; Mc Cafferty, S.; Gansemans, Y.; Joanna Szymańska, K.; Chaitanya Pavani, K.; Catani, J.; Van Nieuwerburgh, F.; Deforce, D.; De Sutter, P.; et al. Bovine Embryo-Secreted Microrna-30c Is a Potential Non-Invasive Biomarker for Hampered Preimplantation Developmental Competence. Front. Genet. 2019, 10, 1–15. [Google Scholar] [CrossRef] [Green Version]
  90. Almiñana, C.; Tsikis, G.; Labas, V.; Uzbekov, R.; da Silveira, J.C.; Bauersachs, S.; Mermillod, P. Deciphering the Oviductal Extracellular Vesicles Content across the Estrous Cycle: Implications for the Gametes-Oviduct Interactions and the Environment of the Potential Embryo. BMC Genom. 2018, 19, 1–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Vidova, V.; Spacil, Z. A Review on Mass Spectrometry-Based Quantitative Proteomics: Targeted and Data Independent Acquisition. Anal. Chim. Acta 2017, 964, 7–23. [Google Scholar] [CrossRef]
  92. Domon, B.; Aebersold, R. Mass Spectrometry and Protein Analysis. Science 2006, 312, 212–217. [Google Scholar] [CrossRef] [Green Version]
  93. Li, X.; Wang, W.; Chen, J. Recent progress in mass spectrometry proteomics for biomedical research. Sci. China Life Sci. 2017, 60, 1093–1113. [Google Scholar] [CrossRef]
  94. Aslam, B.; Basit, M.; Nisar, M.A.; Khurshid, M.; Rasool, M.H. Proteomics: Technologies and Their Applications. J. Chromatogr. Sci. 2017, 55, 182–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Fenn, J.B.; Mann, M.; Meng, C.K.; Wong, S.F.; Whitehouse, C.M. Electrospray ionization for mass spectrometry of large biomolecules. Science 1989, 246, 64–71. [Google Scholar] [CrossRef] [PubMed]
  96. Karas, M.; Hillenkamp, F. Laser desorption ionization of proteins with molecular masses exceeding 10,000 daltons. Anal. Chem. 1988, 60, 2299–2301. [Google Scholar] [CrossRef]
  97. Aebersold, R.; Goodlett, D.R. Mass Spectrometry in Proteomics. Chem. Rev. 2001, 101, 269–295. [Google Scholar] [CrossRef]
  98. Aebersold, R.; Mann, M. Mass spectrometry-based proteomics. Nature 2003, 422, 198–207. [Google Scholar] [CrossRef]
  99. Bantscheff, M.; Schirle, M.; Sweetman, G.; Rick, J.; Kuster, B. Quantitative mass spectrometry in proteomics: A critical review. Anal. Bioanal. Chem. 2007, 389, 1017–1031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Domon, B.; Aebersold, R. Options and considerations when selecting a quantitative proteomics strategy. Nat. Biotechnol. 2010, 28, 710–721. [Google Scholar] [CrossRef]
  101. Mol, P.; Kannegundla, U.; Dey, G.; Gopalakrishnan, L.; Dammalli, M.; Kumar, M.; Patil, A.H.; Basavaraju, M.; Rao, A.; Ramesha, K.P.; et al. Bovine Milk Comparative Proteome Analysis from Early, Mid, and Late Lactation in the Cattle Breed, Malnad Gidda (Bos Indicus). OMICS 2018, 22, 223–235. [Google Scholar] [CrossRef]
  102. Zachut, M.; Sood, P.; Levin, Y.; Moallem, U. Proteomic analysis of preovulatory follicular fluid reveals differentially abundant proteins in less fertile dairy cows. J. Proteom. 2016, 139, 122–129. [Google Scholar] [CrossRef]
  103. Faulkner, S.; Elia, G.; Mullen, M.P.; O’Boyle, P.; Dunn, M.J.; Morris, D. A comparison of the bovine uterine and plasma proteome using iTRAQ proteomics. Proteomics 2012, 12, 2014–2023. [Google Scholar] [CrossRef]
  104. Brown, B.A.; Zeng, X.; Todd, A.R.; Barnes, L.F.; Winstone, J.M.A.; Trinidad, J.C.; Novotny, M.V.; Jarrold, M.F.; Clemmer, D.E. Charge Detection Mass Spectrometry Measurements of Exosomes and other Extracellular Particles Enriched from Bovine Milk. Anal. Chem. 2020, 92, 3285–3292. [Google Scholar] [CrossRef]
  105. Samuel, M.; Chisanga, D.; Liem, M.; Keerthikumar, S.; Anand, S.; Ang, C.S.; Adda, C.G.; Versteegen, E.; Jois, M.; Mathivanan, S. Bovine Milk-Derived Exosomes from Colostrum Are Enriched with Proteins Implicated in Immune Response and Growth. Sci. Rep. 2017, 7, 1–10. [Google Scholar] [CrossRef] [PubMed]
  106. Vaswani, K.; Koh, Y.Q.; Almughlliq, F.B.; Peiris, H.N.; Mitchell, M.D. A method for the isolation and enrichment of purified bovine milk exosomes. Reprod. Biol. 2017, 17, 341–348. [Google Scholar] [CrossRef]
  107. Benmoussa, A.; Gotti, C.; Bourassa, S.; Gilbert, C.; Provost, P. Identification of Protein Markers for Extracellular Vesicle (Ev) Subsets in Cow’s Milk. J. Proteom. 2019, 192, 78–88. [Google Scholar] [CrossRef] [PubMed]
  108. Dalanezi, F.M.; Garcia, H.D.M.; Ferrazza, R.D.A.; Franchi, F.F.; Fontes, P.K.; Castilho, A.C.D.S.; Nogueira, M.F.G.; Schmidt, E.M.D.S.; Sartori, R.; Ferreira, J.C.P. Extracellular vesicles of follicular fluid from heat-stressed cows modify the gene expression of in vitro-matured oocytes. Anim. Reprod. Sci. 2019, 205, 94–104. [Google Scholar] [CrossRef] [PubMed]
  109. Wang, X.; Tian, F.; Chen, C.; Feng, Y.; Sheng, X.; Guo, Y.; Ni, H. Exosome-derived uterine microRNAs isolated from cows with endometritis impede blastocyst development. Reprod. Biol. 2019, 19, 204–209. [Google Scholar] [CrossRef] [PubMed]
  110. Wang, X.; Yao, X.; Xie, T.; Chang, Z.; Guo, Y.; Ni, H. Exosome-derived uterine miR-218 isolated from cows with endometritis regulates the release of cytokines and chemokines. Microb. Biotechnol. 2020, 13, 1103–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Benmoussa, A.; Laugier, J.; Beauparlant, C.J.; Lambert, M.; Droit, A.; Provost, P. Complexity of the Microrna Transcriptome of Cow Milk and Milk-Derived Extracellular Vesicles Isolated Via Differential Ultracentrifugation. J. Dairy Sci. 2020, 103, 16–29. [Google Scholar] [CrossRef]
  112. Ma, S.; Tong, C.; Ibeagha-Awemu, E.M.; Zhao, X. Identification and Characterization of Differentially Expressed Exosomal Micrornas in Bovine Milk Infected with Staphylococcus Aureus. BMC Genom. 2019, 20, 1–13. [Google Scholar] [CrossRef] [Green Version]
  113. Zhao, G.; Guo, S.; Jiang, K.; Zhang, T.; Wu, H.; Qiu, C.; Deng, G. MiRNA profiling of plasma-derived exosomes from dairy cows during gestation. Theriogenology 2019, 130, 89–98. [Google Scholar] [CrossRef]
  114. McDougall, S.; Macaulay, R.; Compton, C. Association between endometritis diagnosis using a novel intravaginal device and reproductive performance in dairy cattle. Anim. Reprod. Sci. 2007, 99, 9–23. [Google Scholar] [CrossRef]
  115. Paiano, R.B.; Gonçalves, C.G.P.; Mendes, J.P.G.; Bonilla, J.; Birgel, D.B.; Junior, E.H.B. Comparative biochemical profiles, production and reproduction status of the post-partum dairy cows with and without purulent vaginal discharge. Reprod. Domest. Anim. 2019, 54, 1188–1194. [Google Scholar] [CrossRef]
  116. Anjo, S.I.; Santa, C.; Manadas, B. Swath-Ms as a Tool for Biomarker Discovery: From Basic Research to Clinical Applications. Proteomics 2017, 17, 3–4. [Google Scholar] [CrossRef]
  117. Ghodasara, P.; Sadowski, P.; Satake, N.; Kopp, S.; Mills, P.C. Clinical Veterinary Proteomics: Techniques and Approaches to Decipher the Animal Plasma Proteome. Vet. J. 2017, 230, 6–12. [Google Scholar] [CrossRef] [PubMed]
  118. Ludwig, C.; Gillet, L.; Rosenberger, G.; Amon, S.; Collins, B.C.; Aebersold, R. Data-Independent Acquisition-Based Swath—Ms for Quantitative Proteomics: A Tutorial. Mol. Syst. Biol. 2018, 14, 1–23. [Google Scholar] [CrossRef] [PubMed]
  119. Gillet, L.C.; Navarro, P.; Tate, S.; Rost, H.; Selevsek, N.; Reiter, L.; Bonner, R.; Aebersold, R. Targeted Data Extraction of the Ms/Ms Spectra Generated by Data-Independent Acquisition: A New Concept for Consistent and Accurate Proteome Analysis. Mol. Cell Proteom 2012, 11, O111-016717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Menon, R.; Dixon, C.L.; Sheller-Miller, S.; Fortunato, S.J.; Saade, G.R.; Palma, C.; Lai, A.; Guanzon, D.; Salomon, C. Quantitative Proteomics by SWATH-MS of Maternal Plasma Exosomes Determine Pathways Associated With Term and Preterm Birth. Endocrinol. 2019, 160, 639–650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Chutipongtanate, S.; Greis, K.D. Multiplex Biomarker Screening Assay for Urinary Extracellular Vesicles Study: A Targeted Label-Free Proteomic Approach. Sci. Rep. 2018, 8, 15039. [Google Scholar] [CrossRef]
  122. Guan, S.; Taylor, P.P.; Han, Z.; Moran, M.F.; Ma, B. Data Dependent–Independent Acquisition (DDIA) Proteomics. J. Proteome Res. 2020, 19, 3230–3237. [Google Scholar] [CrossRef]
  123. Schubert, O.T.; Gillet, L.C.; Collins, B.C.; Navarro, P.; Rosenberger, G.; Wolski, W.E.; Lam, H.; Amodei, D.; Mallick, P.; MacLean, B.; et al. Building high-quality assay libraries for targeted analysis of SWATH MS data. Nat. Protoc. 2015, 10, 426–441. [Google Scholar] [CrossRef] [PubMed]
  124. Collins, B.C.; Hunter, C.L.; Liu, Y.; Schilling, B.; Rosenberger, G.; Bader, S.L.; Chan, D.W.; Gibson, B.W.; Gingras, A.-C.; Held, J.M.; et al. Multi-laboratory assessment of reproducibility, qualitative and quantitative performance of SWATH-mass spectrometry. Nat. Commun. 2017, 8, 291. [Google Scholar] [CrossRef] [PubMed]
  125. Selevsek, N.; Chang, C.-Y.; Gillet, L.C.; Navarro, P.; Bernhardt, O.M.; Reiter, L.; Cheng, L.-Y.; Vitek, O.; Aebersold, R. Reproducible and Consistent Quantification of the Saccharomyces cerevisiae Proteome by SWATH-mass spectrometry. Mol. Cell. Proteom. 2015, 14, 739–749. [Google Scholar] [CrossRef] [Green Version]
  126. Baranyai, T.; Herczeg, K.; Onódi, Z.; Voszka, I.; Módos, K.; Marton, N.; Nagy, G.; Mäger, I.; Wood, M.J.; El Andaloussi, S.; et al. Isolation of Exosomes from Blood Plasma: Qualitative and Quantitative Comparison of Ultracentrifugation and Size Exclusion Chromatography Methods. PLoS ONE 2015, 10, e0145686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Diaz, G.; Bridges, C.; Lucas, M.; Cheng, Y.; Schorey, J.S.; Dobos, K.M.; Kruh-Garcia, N.A. Protein Digestion, Ultrafiltration, and Size Exclusion Chromatography to Optimize the Isolation of Exosomes from Human Blood Plasma and Serum. J. Vis. Exp. 2018, 134, 1–6. [Google Scholar] [CrossRef] [PubMed]
  128. Koh, Y.Q.; Almughlliq, F.B.; Vaswani, K.; Peiris, H.N.; Mitchell, M.D. Exosome Enrichment by Ultracentrifugation and Size Exclusion Chromatography. Front. Biosci. (Landmark Ed.) 2018, 23, 865–874. [Google Scholar] [PubMed] [Green Version]
  129. Konoshenko, M.Y.; Lekchnov, E.A.; Vlassov, A.V.; Laktionov, P.P. Isolation of Extracellular Vesicles: General Methodologies and Latest Trends. BioMed Res. Int. 2018, 2018, 1–27. [Google Scholar] [CrossRef] [PubMed]
  130. Martins, T.S.; Catita, J.; Rosa, I.M.; Silva, O.A.B.D.C.E.; Henriques, A.G. Exosome isolation from distinct biofluids using precipitation and column-based approaches. PLoS ONE 2018, 13, e0198820. [Google Scholar] [CrossRef] [Green Version]
  131. Pietrowska, M.; Wlosowicz, A.; Gawin, M.; Widlak, P. Ms-Based Proteomic Analysis of Serum and Plasma: Problem of High Abundant Components and Lights and Shadows of Albumin Removal. Emerg. Sample Treat. Proteom. 2019, 1073, 57–76. [Google Scholar]
Figure 1. Routes of exosomal formation and release from the cell. The Golgi apparatus (1) transports and modifies proteins received from the endoplasmic reticulum (ER). Mature proteins and proproteins are transferred from the Golgi to endosomes via transport vesicles (2a and 3). Early endosomes go on to form late endosomes/multivesicular bodies (MVBs) (4 and 5), which are composed of intraluminal vesicles (ILVs) formed from the inward budding of the endosomal membrane during the maturation process. Endosomal sorting complex required for transport (ESCRT) proteins are involved in this process and are found in ILV cargo. MVBs fuse with the plasma membrane of the cell to release their contents into the extracellular milieu; extracellular vesicles (EVs) (6). EVs are taken up by the cell via endocytosis or phagocytosis (2b) and transported to endosomal compartments and lysosomes for processing [37].
Figure 1. Routes of exosomal formation and release from the cell. The Golgi apparatus (1) transports and modifies proteins received from the endoplasmic reticulum (ER). Mature proteins and proproteins are transferred from the Golgi to endosomes via transport vesicles (2a and 3). Early endosomes go on to form late endosomes/multivesicular bodies (MVBs) (4 and 5), which are composed of intraluminal vesicles (ILVs) formed from the inward budding of the endosomal membrane during the maturation process. Endosomal sorting complex required for transport (ESCRT) proteins are involved in this process and are found in ILV cargo. MVBs fuse with the plasma membrane of the cell to release their contents into the extracellular milieu; extracellular vesicles (EVs) (6). EVs are taken up by the cell via endocytosis or phagocytosis (2b) and transported to endosomal compartments and lysosomes for processing [37].
Ijms 22 02024 g001
Figure 2. Blended model of reproduction and inflammation: Arachidonic Acid (AA)/Eicosanoid Pathway. Fatty acid cyclooxygenase 1/2 (Cox 1/2) converts AA to downstream effector molecules (Prostanoids and Prostaglandins (PGs)) following inflammatory stimuli. Interferon-tau (IFN-τ) produced by the conceptus inhibits Oxytocin receptor (Oxtr) expression and prevents luteolysis of luteinized granulosa cells to maintain progesterone secretion. IFN-τ stimulates PGE2 production in the endometrium, resulting in structural and functional changes required for pregnancy recognition. In vitro studies show altered expression of PGF and PGE2 when exposed to inflammatory stimuli, which in turn may compromise events leading to successful establishment of pregnancy. Nonsteroidal anti-inflammatory drugs (NSAIDs) target the PG inflammatory cascade by inhibiting Cox2 expression and reducing production of PGH2 and associated inflammatory mediators.
Figure 2. Blended model of reproduction and inflammation: Arachidonic Acid (AA)/Eicosanoid Pathway. Fatty acid cyclooxygenase 1/2 (Cox 1/2) converts AA to downstream effector molecules (Prostanoids and Prostaglandins (PGs)) following inflammatory stimuli. Interferon-tau (IFN-τ) produced by the conceptus inhibits Oxytocin receptor (Oxtr) expression and prevents luteolysis of luteinized granulosa cells to maintain progesterone secretion. IFN-τ stimulates PGE2 production in the endometrium, resulting in structural and functional changes required for pregnancy recognition. In vitro studies show altered expression of PGF and PGE2 when exposed to inflammatory stimuli, which in turn may compromise events leading to successful establishment of pregnancy. Nonsteroidal anti-inflammatory drugs (NSAIDs) target the PG inflammatory cascade by inhibiting Cox2 expression and reducing production of PGH2 and associated inflammatory mediators.
Ijms 22 02024 g002
Table 1. Summary of knowledge relating to exosomes and dairy cow reproduction.
Table 1. Summary of knowledge relating to exosomes and dairy cow reproduction.
KnownNot KnownFuture Direction
Characterization of plasma exosomes derived from high- and low-fertility dairy cows [16].‘Gold standard’ for exosome isolation is still a matter of contention.Further optimization of exosomal isolation protocols specific to downstream application.
Characterization of bovine milk exosomes [16].--
Established proteome profile of plasma exosomes derived from high- and low-fertility dairy cows [4,10].Quantitative proteomic profile of exosomal cargo in circulating bovine exosomes.SWATH-MS proteomic analysis of circulating exosomes in high- and low-fertility dairy cows to confirm quantitative differences and identify biomarker candidates related to good/poor reproductive outcomes.
Established proteome profile of bovine exosomes derived from milk, follicular fluid and uterine flushings [47,48,105,107,108].Comprehensive quantitative proteomic profile of exosomes derived from bovine milk, follicular fluid and uterine flushings.SWATH-MS proteomic analyses of exosomes derived from these biological fluid types to obtain a more complete understanding of the connection between physiological processes involved in dairy cow reproduction.
Characterization of bovine endometrial inflammation via in vitro inflammatory model utilizing bovine endometrial epithelial (bEEL) and stromal cells (bCSC) [58]. Exosomes derived from cows with uterine infection were found to decrease PGF production in bEEL, but not bCSC cell lines [51]. Exosomes derived from cows at high- or low-risk of metabolic dysfunction differentially regulate eicosanoid gene expression in bEEL and bCSC cell lines [50].In vitro studies utilizing novel protein biomarkers associated with healthy/aberrant reproduction.Pathway analysis of potential biomarkers identified in protein studies and ongoing in vitro experiments to confirm biological function/impact of candidate biomarkers on eicosanoid gene and protein expression.
Exosome-derived uterine miRNAs from dairy cows are involved in blastocyst development and regulation of cytokines and chemokines [109,110].Effect of miRNA knockdown on the function in relation to regulation of reproductive processes.miRNA knockdown/knockout studies to confirm involvement of miRNA on the regulation of bovine reproductive processes.
Established miRNA profiles of bovine plasma- and milk-derived exosomes [111,112,113].Comparative studies relating to exosomal miRNA profiles of high- and low-fertility dairy cattle.Perform qualitative and quantitative analysis of exosomal miRNA in high- and low-fertility groups.
Immune challenges are associated with poor reproductive outcomes in dairy cows [41,62,114,115].Relationship between immune status and poor reproductive outcomes needs further clarification.Continuing studies on inflammatory mediators and their relationship to reproductive processes.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Turner, N.; Abeysinghe, P.; Sadowski, P.; Mitchell, M.D. Exosomal Cargo May Hold the Key to Improving Reproductive Outcomes in Dairy Cows. Int. J. Mol. Sci. 2021, 22, 2024. https://doi.org/10.3390/ijms22042024

AMA Style

Turner N, Abeysinghe P, Sadowski P, Mitchell MD. Exosomal Cargo May Hold the Key to Improving Reproductive Outcomes in Dairy Cows. International Journal of Molecular Sciences. 2021; 22(4):2024. https://doi.org/10.3390/ijms22042024

Chicago/Turabian Style

Turner, Natalie, Pevindu Abeysinghe, Pawel Sadowski, and Murray D. Mitchell. 2021. "Exosomal Cargo May Hold the Key to Improving Reproductive Outcomes in Dairy Cows" International Journal of Molecular Sciences 22, no. 4: 2024. https://doi.org/10.3390/ijms22042024

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop