Skip to main content

Seasonal environmental fluctuations alter the transcriptome dynamics of oocytes and granulosa cells in beef cows

Abstract

Background

Examining the mechanistic cellular responses to heat stress could aid in addressing the increasing prevalence of decreased fertility due to elevated ambient temperatures. Here, we aimed to study the differential responses of oocytes and granulosa cells to thermal fluctuations due to seasonal differences. Dry beef cows (n = 10) were housed together, synchronized and subjected to a stimulation protocol to induce follicular growth before ovum pick-up (OPU). Two OPU’s were conducted (summer and winter) to collect cumulus-oocyte-complexes (COCs) and granulosa cells. In addition, rectal temperatures and circulating blood samples were collected during OPU. Oocytes were separated from the adherent cumulus cells, and granulosa cells were isolated from the collected OPU fluid. RNA was extracted from pools of oocytes and granulosa cells, followed by library preparation and RNA-sequencing. Blood samples were further processed for the isolation of plasma and leukocytes. The transcript abundance of HSP70 and HSP90 in leukocytes was evaluated using RT-qPCR, and plasma cortisol levels were evaluated by immunoassay. Environmental data were collected daily for three weeks before each OPU session. Data were analyzed using MIXED, Glimmix or GENMOD procedures of SAS, according to each variable distribution.

Results

Air temperatures (27.5 °C vs. 11.5 °C), average max air temperatures (33.7 °C vs. 16.9 °C), and temperature-humidity indexes, THI (79.16 vs. 53.39) were shown to contrast significantly comparing both the summer and winter seasons, respectively. Rectal temperatures (Summer: 39.2 ± 0.2 °C; Winter: 38.8 ± 0.2 °C) and leukocyte HSP70 transcript abundance (Summer: 4.18 ± 0.47 arbitrary units; Winter: 2.69 ± 0.66 arbitrary units) were shown to increase in the summer compared to the winter. No visual differences persisted in HSP90 transcript abundance in leukocytes and plasma cortisol concentrations during seasonal changes. Additionally, during the summer, 446 and 940 transcripts were up and downregulated in oocytes, while 1083 and 1126 transcripts were up and downregulated in the corresponding granulosa cells, respectively (Fold Change ≤ -2 or ≥ 2 and FDR ≤ 0.05). Downregulated transcripts in the oocytes were found to be involved in ECM-receptor interaction and focal adhesion pathways, while the upregulated transcripts were involved in protein digestion and absorption, ABC transporters, and oocyte meiosis pathways. Downregulated transcripts in the granulosa cells were shown to be involved in cell adhesion molecules, chemokine signaling, and cytokine-cytokine receptor interaction pathways, while those upregulated transcripts were involved in protein processing and metabolic pathways.

Conclusion

In conclusion, seasonal changes dramatically alter the gene expression profiles of oocytes and granulosa cells in beef cows, which may in part explain the seasonal discrepancies in pregnancy success rates during diverging climatic weather conditions.

Background

With the rise of climate change becoming more prevalent over the years, it has been postulated that the duration and severity of heat stress will continually increase, posing detrimental impacts to the livestock industry, notably the beef and dairy sectors [1]. Excessive heat loads on animals negatively impact animal welfare and performance, with evident reductions in reproductive efficiencies, as one of the leading heat stress-related impacts affecting livestock productivity [2]. Additionally, it is well-documented that heat stress negatively impacts the reproductive performance of dairy and beef cattle through alterations in follicular development, oocyte maturation, and granulosa cells’ function [3, 4]. Therefore, understanding the molecular changes in oocytes and follicular cells associated with environmental heat stress in beef cows could provide a unique opportunity to identify molecular targets for future managerial interventions and the development of therapeutic strategies.

Seasonal studies in cattle have shown that elevated temperatures compromise oocyte development [5]. Rocha et al. [6] observed the effects of environmental temperatures and humidity on the quality and developmental capacity of bovine oocytes from Holstein cows [6]. Results from this study showed that a higher percentage of morphologically normal oocytes were produced during the cooler season than during the warmer season, while the percentage of fertilized oocytes that developed to the 2-cell, 8-cell, and morula stages, were also greater during the cooler season. Moreover, previous studies have duly indicated that altered oocyte developmental competence from direct exposure to heat stress also negatively hampers their mitochondrial function by impacting ATP levels and redox regulation [7].Furthermore,, heat stress also has negative impacts on the function of granulosa cells. Disturbances in granulosa cells’ quality and impairments to their proliferation capacity may indirectly affect follicular development and oocyte maturation, resulting in impaired embryo development and subsequent failures in pregnancy establishment [4]. Furthermore, in mice, it was shown that the exposure to heat stress increased the proportion of granulosa cells undergoing apoptosis and the number of atretic follicles, while reducing the intrafollicular aromatase activity, as well as circulating estradiol concentrations [8]. Additional studies are required to examine the molecular mechanisms underlying the response of oocytes and granulosa cells to heat stress, specifically in beef cattle.

A recent study from our group [9] investigated the encapsulated microRNA cargo of extracellular vesicles (EVs) derived from the follicular fluid of beef cows in response to seasonal environmental changes. In that study, several miRNAs were differentially expressed in EVs derived from the summer versus the winter, with precise candidates like miR-10a, miR-10b, miR-184, miR-19b, and miR-452 upregulated and miR-1246, miR-199b, and miR-370 downregulated during summer compared to the winter groups. Interestingly, those DE-miRNAs were found to be involved in the regulation of signaling pathways including WNT-, p53-, mTOR-, Hippo, and FoxO-signaling pathways, which are the dominant regulatory pathways involved in mammalian follicular development. As follicular fluid EV molecular cargos are direct determinants and a reflection of the physiology of the cells amidst the follicular microenvironment including cumulus cells, granulosa cells, and theca cells, the results of our previous study led us to hypothesize that the oocytes and the surrounding granulosa cells respond to environmental seasonal fluctuations in heat stress via altering their transcriptome profile. Therefore, in the present study, we aimed to investigate the transcriptome dynamics of GV-stage oocytes and granulosa cells, determining their response to fluctuating environmental temperatures during summer and winter seasons.

Materials and methods

Animals

This experiment was conducted at the UF/IFAS North Florida Research and Education Center in Marianna, FL, and was approved by the Institutional Animal Care and Use Committee (IACUC ID: IACUC202200000706). Eleven non-pregnant and non-lactating, multiparous Bos taurus cows crossbreed were clinically examined via transrectal ultrasound to evaluate their reproductive tract and cyclicity status. Only animals without clinical abnormalities showing at least one corpus luteum were enrolled in the study. Cows remained together for the duration of the experiment (February-August) and were maintained in outdoor pens with Bahia grass and fed bermudagrass hay to meet the nutritional requirements of mature cows with ad libitum access to water and mineralized salt during the duration of the study. One cow died before the second OPU session and therefore, was completely removed from the study.

Synchronization, follicular stimulation, and ovum pick-up

Cows were subjected to a synchronization and FSH-based stimulation protocol to induce follicular growth as indicated in the hormonal experimental design outlined in Fig. 1. On day 0, an intravaginal P4 device (Eazi-Breed™ CIDR®; 1.38 gr P4; Zoetis Animal Health; Kalamazoo, MI, USA) was inserted and an intramuscular injection of a GnRH analog (100 µg of synthetic gonadorelin hydrochloride; Factrel; Zoetis Animal Health; Kalamazoo, MI, USA) was administrated. Follicular growth was stimulated by administering exogenous FSH (porcine pituitary-derived follicle-stimulating hormone; Folltropin®; Vetoquinol; Canada). Briefly, on day 3, 105 IU of FSH were administrated intramuscularly twice, once in the morning (7 am) and once in the afternoon (6 pm). Next, on day 4, one injection of 70 IU of FSH was administered in the morning (7 am). Finally, the CIDR was removed on day 5 (morning), and rectal temperature was collected. Transrectal ultrasounds were then conducted to assess the number of antral follicles and the presence of a CL. OPU was performed as previously reported [9] using a real-time B-mode ultrasound scanner (Mindray 2200; Mindray Bio-Medical Electronics, Shenzhen, China) equipped with a 5-MHz micro-convex transducer (Mindray model 65C15EAV, Mindray Bio-Medical Electronics, Shenzhen, China) and coupled to a follicular aspiration guide (WTA, São Paulo, Brazil) and a stainless-steel guide. The follicular puncture was performed using a disposable 18 G hypodermic needle connected to a 50-mL conical tube via a suitable silicon tubing system (WTA). The pressure for aspiration was maintained using a vacuum pump (WTA model BV-003, WTA) with negative pressure adjusted between 60 and 80 mmHg. Only follicles from ~ 4 mm to ≤ 8 mm were aspirated. Follicles ≥ to 9 mm were starting a dominance phase and were not aspirated. After the OPU of both ovaries, the aspiration system was replaced with a new one before conducting OPU for the next cow. OPUs from winter and summer were conducted in February and August, respectively by the same operator.

Fig. 1
figure 1

Hormonal protocol used in the present study. Briefly, a 5-day CIDR and FSH synchronization and stimulation protocol was used to induce follicular growth before each OPU session (Winter and Summer). Created with BioRender.com

Sample collections

Collection of cumulus-oocyte-complexes and oocyte isolation

Following OPU procedures for each animal, COCs were allowed to settle to the bottom of the collection tube. The uppermost aqueous fluid above the pelleted aspirate was then collected with a pipette and transferred to a new tube. Following searching the dense collection of cellular debris and tissue remnants, COCs were washed three times with holding media (ABT complete flush; ABT 360, LLC; Pullman, WA, USA). Grading of COCs was performed based on the criteria set by the International Embryo Technology Society [10]. Grade 1: >5 layers of even cumulus cells with even cytoplasm; Grade 2: three to five layers of cumulus cells, mostly even distributed, and even cytoplasm; Grade 3: < 3 layers of dense compact cumulus cells, often uneven, and abnormally small oocytes with clear, granular, and uneven cytoplasm; Grade 4: degenerated. Grade 3 and 4 COCs were discarded and not included in further analyses of this study.

Collected grades 1 and 2 COCs were immediately subjected to enzymatic and mechanical efforts to remove adhered cumulus cells using 10,000 units/mL hyaluronidase (Sigma H-4272) and a high-speed vortex. One ml of holding medium was added to each tube, and the contents were transferred to a separate petri dish. Denuded oocytes (germinal vesicle stage, GV) were then isolated and washed twice in PBS. Every oocyte was closely checked for the presence of somatic cells under a stereomicroscope. Finally, the oocytes were transferred individually to a new tube with minimal volume of PBS and immediately snap-frozen in liquid nitrogen. All samples were stored at -80oC until further analysis.

Collection of granulosa cells

For individual animals, the uppermost aqueous phase of the OPU collection fluid was transferred to a new 15 ml falcon tube and centrifuged at 750 ×g for 5 min to pelletize the cells. Granulosa cells were then immediately snap-frozen and stored at -80oC until further use.

Collection of plasma samples

At the time of each OPU, blood samples were collected using EDTA blood collection tubes (K2 EDTA BD Vacutainer®; Ref. 366643; BD Vacutainer; United Kingdom). OPU session happened in the morning period for both, summer and winter collections. Collection tubes containing the blood samples were centrifuged at 3000 ×g for 15 min to separate the plasma from the blood cells. The plasma was pipetted into microtubes and stored at -80oC for downstream analysis.

Leukocytes isolation

The white blood cell layer resulting from harvesting the plasma was then used to harvest the buffy coat. Briefly, the white layer was transferred into a 15 ml falcon tube using a sterile pipette and diluted in 3x the volume (~ 6 ml) of distilled water. Cells were mechanically suspended via pipetting, followed by centrifugation at 500×g for 15 min at 18oC. The supernatant was removed, and the pellets were scraped against a rack to unstick the cells. This step was repeated until the pellet was cleared of all red blood cells (2–3 times). Finally, the resuspended cells were transferred to a 2 ml microtube and centrifuged at 4000 ×g for 10 min. Without disturbing the cell pellet, the supernatant was removed, and the ensuing leukocytes were then immediately snap-frozen and stored at -80oC for downstream application.

Climatic conditions

Data for climatic conditions were collected daily for three weeks before each OPU session using the Florida Automated Weather Network (FAWN) [11]. The following weather measurements were taken from FAWN: temperature (average and maximum) and relative humidity. The temperature humidity index (THI) was calculated using the following equation: THI = (1.8 × T + 32) - [(0.55–0.0055 × RH) × (1.8 × T − 26)]. Where T = air temperature (°C) and RH = relative humidity (%) [12].

Plasma cortisol assay

Circulating plasma cortisol concentrations were quantified using the automated IMMULITE® 2000 Xpi cortisol Immunoassay kit (Cat. No. L2KCO2, Siemens Healthcare, CA, USA). Briefly, per the manufacturer’s recommendations, the cortisol kit was adjusted for each lot number with a calibration range of 10 to 500 ng/mL. Commercially available tri-level (26.8, 149, and 254 ng/mL) quality control samples (Lyphocheck Immunoassay Plus Control, Cat. No. 370, Bio-Rad, Hercules, CA, USA) were run daily before the experimental samples. Quality control samples were considered to pass if the coefficient of variation (CV) was below 10% for each level and the replicates were within the manufacturer’s reported range. Before analysis, samples were thawed at room temperature and run in duplicates. An internal calculation of quantification was reported for each duplicate, and these values were averaged to determine the CV. All concentrations reported have a CV of less than 10%.

Total RNA extraction, reverse transcription, and quantitative real-time PCR of leukocytes

Total RNA was extracted from leukocytes using the PureLink™ RNA Mini Kit (Cat. No. 12183025; Thermo Fisher Scientific™; Austin, TX, USA) according to the manufacturer’s instruction. Samples were homogenized in 600 µL of lysis buffer containing 1% mercaptoethanol. The “On-Column PureLink® DNase Treatment Protocol” from the PureLink™ DNase Set (Cat. No. 12185010; Thermo Fisher Scientific™) was performed on individual samples to remove DNA contaminants. RNA yield and purity were analyzed using the NanoDrop™ One Spectrophotometer (Thermo Fisher Scientific™) with criteria set that the yield (> 20 ng/µL) and the purity (absorbance 260/280 ratio ~ 2). Cows containing valid samples from summer and winter, were keep for further analysis. Samples were then stored at -80 °C until further use.

RNA samples (n = 7 in summer and n = 7 in winter) were reverse transcribed to cDNA using the High-Capacity cDNA Reverse Transcription Kit (Cat. No. 4368814; Thermo Fisher Scientific™) following the manufacturer’s instruction. Briefly, 500 ng of RNA and ensuing reagents were combined and subjected to incubation in a Bio-Rad C1000 Touch™ Thermal Cycler at an initial temperature of 25oC for 10 min, followed by 37oC for 120 min, 85oC for 5 min, and then 4oC. The final output volume of 20 µL of cDNA was then diluted 1:80 with DEPC-treated water and stored at -20 °C for further qRT-PCR reactions.

Primers used for quantitative real-time PCR (qPCR) are indicated in Supplementary Table 1. The characteristics of the primers were checked in The OligoAnalyzer™ Tool software, (Integrated DNA Technologies, Biodynamics, https://www.idtdna.com/pages/tools/oligoanalyzer?returnurl=%2Fcalc%2Fanalyzer), while the specificity was compared by BLAST1 (NCBI, http://blast.ncbi.nlm.nih.gov). each primer pair were previously validated to establish primer pair efficiency (standard curves) and optimal primer concentration (concentration test). For all reactions (target and reference genes), a master mix containing SYBR Green (Cat. No. 1725274; SsoAdvanced Universal SYBR Green Supermix; Bio-Rad), forward and reverse primers, and DEPC-treated water were prepared in a 2 ml microtube. qPCR reactions were conducted in triplicates and prepared in a 96-well PCR plate (Cat. No. HSP9601; Bio-Rad) containing 16 µL of the master mix and 4 µL of the diluted cDNA samples. No template controls (NTC) without cDNA were run in each PCR plate. The Bio-Rad CFX software was used to run the protocol as follows: denaturation at 95oC for 3 min, followed by 40 cycles of 95oC for 10 s, and 60oC for 30 s, and finally, annealing/extension and plate read at 65oC for 5 s and then melting curve analysis at 95oC + 0.5oC. No amplification was detected in NTC. RefFinder [13, 14] was used to determine the stability of the reference genes, and according to their Cq, ACT-B, and RPL15 were the most stable reference genes. Analysis to determine the relative expression of the genes of interest was done using the PFAFFL equation [15], considering each primer’s efficiency and following normalization using the geometric mean of the selected reference genes (ACT-B and RPL15). qPCR products from reactions containing target and reference genes primers were submitted to agarose gel electrophoresis and sequencing and identities were confirmed.

Preparation of RNA from oocytes and granulosa cells for sequencing

Total RNA extraction of oocytes was performed after pooling 2–4 oocytes from a single cow and using the Arcturus™ PicoPure™ RNA (Thermo Fisher Scientific™) according to the manufacturer’s instruction Total RNA from granulosa cells was extracted using the miRNeasy mini kit (Qiagen, Hilden, Germany). For both types of samples (oocytes and granuloma cells), on-column DNA digestion was performed using the RNase-Free DNase Set (Qiagen) to remove genomic DNA contaminants. The concentration and integrity of the isolated samples were then assessed with NanoDrop 8000 spectrophotometer (Thermo Fisher Scientific™) and Agilent 2100 Bioanalyzer (Agilent Technologies, CA), respectively. RNA samples were pooled from two individual animals to create five biological replicates (n = 2 cows/pool, 5 pools per season) for both oocytes and granulosa cells, collected during the summer and winter. RNA samples were stored at -80 °C until sequencing.

Library preparation and RNA-sequencing

For library preparation, messenger RNA was purified from total RNA using poly-T oligo-attached magnetic beads. After fragmentation, the first strand of cDNA was synthesized using random hexamer primers, followed by the second strand of cDNA synthesis. The library was ready after the end repair, A-tailing, adapter ligation, size selection, amplification, and purification. The libraries were checked with a Qubit DNA HS Assay Kit in a Qubit 2.0 Fluorometer (Thermo Fisher Scientific™) for concentration, qPCR for quantification, and a bioanalyzer (Agilent Technologies) for the size distribution. Quantified libraries were pooled and sequenced using a NovaSeq6000 sequencing instrument (Illumina, Inc., San Diego, CA, USA) as paired end reads (150 bases).

Sequencing data analysis

FASTQ files were generated for each sample using the software bcl2fastq (Illumina Inc., San Diego, CA), and their quality was checked using the FastQC tool version 0.11.9. Data were analyzed using the CLC Genomics Workbench, version 21 (Qiagen). Raw sequencing reads were trimmed based on quality score (Q-score > 30), ambiguous nucleotides (maximum two nucleotides allowed), read length (≥ 15 nucleotides), and after removing adapter sequences. One replicate from oocyte/summer group didn’t pass the quality control check. Therefore, we decided to exclude this replicate and its corresponding oocyte/winter and granulosa cell replicates from further analysis. Reads were mapped to the bovine reference genome (ARS-UCD1.3) applying the default software parameters. Data were normalized using the trimmed mean of the M-values normalization method (TMM normalization) [16] and presented as transcripts per million (TPM). The expression threshold was determined by the zFPKM method using the zFPKM R package v.1.16.0 [17]. Genes with zFPKM > − 3 in all replicates were considered expressed. Differential expression analysis was done using the Differential Expression tool based on a negative binomial Generalized Linear Model (GLM) function. Differentially expressed genes (DEGs) were filtered based on fold change (FC ≥ 2) and the P-adjusted value (FDR < 0.05) [18]. The raw FASTQ files and processed CSV files have been deposited into NCBI’s Gene Expression Omnibus (GEO) with the accession numbers (GSE235170, GSE235171) for the oocytes and granulosa cells, respectively.

After the identification of the DEGs in each cell type separately, a comparative analysis has been conducted to elucidate the unique and shared molecular responses of oocytes and granulosa cells to elevated environmental temperatures. Specifically, we focused on genes that were commonly dysregulated in both cell types during the summer followed by pathway enrichment analysis to identify the biological pathways impacted by these DEGs.

Pathways and ontological classification analysis

The DEGs were submitted to the Database for Annotation, Visualization, and Integrated Discovery (DAVID) Bioinformatics web tool v. 2021 [19] for pathways and ontological classification enrichment analysis. Pathways and biological processes (BP) were determined from the KEGG database [20] and GOTERM_BP_DIRECT annotation set, respectively.

Statistical analysis

Rectal temperature, plasma cortisol concentration, and HSP70 and HSP90 transcript abundance in leukocytes were analyzed using the MIXED procedure of SAS version 9.3 (SAS/STAT®, SAS Inst. Inc., Cary, NC) with cow as a random effect. The number of follicles and oocytes were analyzed by negative binomial regression analyses using the GENMOD procedure. The proportion of grade 1, 2, 3, and 4 oocytes were analyzed by ANOVA using the GLIMMIX procedure with logit function. Square-root transformation was applied when residuals were not normally distributed according to the Shapiro-Wilk and Kolmogorov-Smirnov tests. For all models, cow was considered the experimental unit, and data are presented as mean ± standard error of the mean. We set the significance level for rejecting the null hypothesis to P ≤ 0.05 and considered trends significant at 0.05 ≤ P ≤ 0.10.

Results

Climatic conditions

As expected, the average air temperature (27.5 °C vs. 11.5 °C), average maximum air temperature (33.7 °C vs. 16.9 °C), and relative humidity (82.3% vs. 83.5%) during the summer and winter, respectively, differed considerably (Fig. 2). After using the equation to calculate the THI for the 3 weeks prior to each sample collection, the summer estimate was 79.2, while the winter was 53.4 (Fig. 2; Table 1). Accordingly, the Livestock Safety Weather Index [21], a common resource used to classify the intensity of the THI values, recognizes four primary categories: normal, alert, danger, and emergency. Based on this criterion, over the three weeks preceding the OPU collections, the winter had 100.0% normal THI days, while the summer had 59.1% of the days considered as ‘alert’ and 40.9% considered as ‘danger’ THI thresholds (Table 1).

Fig. 2
figure 2

Climatic conditions were collected for twenty-one days before each OPU session. A) Average air temperature, B) Relative humidity, C) Temperature humidity index (THI). Data were collected using the Florida Automated Weather Network (FAWN; https://fawn.ifas.ufl.edu/data/reports/)

Table 1 Descriptive statistics for the temperature humidity index (THI) average values for 21 days before each OPU session

Rectal temperature and plasma cortisol concentration

Average rectal temperatures were higher (P = 0.03) in the summer (39.2 ± 0.2 °C) than in the winter (38.8 ± 0.2 °C; Fig. 3A). However, plasma cortisol concentrations did not differ when comparing collections from the summer (29.4 ± 3.0 ng/mL) and winter (34.4 ± 4.2 ng/mL) seasons (Fig. 3B).

Fig. 3
figure 3

Mean ± SEM of rectal temperature (A), plasma cortisol concentration (B), and HSP70 (C) and HSP90 (D) transcript abundance from beef cows during the summer and winter (n = 11). *** Denotates statistical differences when P ≤ 0.05 and * denotates statistical tendencies when P ≥ 0.05 still P < 0.1

HSP70 and HSP90 transcript abundance in leukocytes

Quantitative analysis of HSP70 and HSP90 in leukocytes isolated from cows during the summer and winter seasons revealed that HSP70 tended to have a higher abundance during the summer season, with no difference in the expression of HSP90 (Fig. 3C and D).

Follicular observations and oocyte recovery

No significant difference in the total number of follicles (summer 23.9 ± 3.9 vs. winter 18.9 ± 4.8) and the total number of oocytes recovered (summer 13.0 ± 3.6 vs. winter 9.0 ± 1.6) between seasonal collections was observed (Table 2). However, oocyte quality differed when comparing the seasons. In summer, both the number (summer 1.09 ± 0.5 vs. winter 6.4 ± 1.7; P = < 0.01) and the proportion (summer 7.1% ± 2.5 vs. winter 57.8% ± 12.0; P = < 0.01) of grade 1 oocytes decreased, while the number (summer 4.1 ± 1.1 vs. winter 1.2 ± 0.7; P = 0.09) and the proportion (summer 30.5% ± 5.5 vs. winter 12.3% ± 5.2; P = 0.04) of grade 4 oocytes increased or tended to increased. The number of grade 2 and grade 3 oocytes did not differ significantly between seasonal collections, however, the proportion of grade 2 (summer 26.3% ± 8.4 vs. winter 11.4% ± 5.6; P = 0.05) and grade 3 (summer 36.1% ± 5.4 vs. winter 8.3% ± 8.3; P = < 0.01) oocytes was higher from collections during the summer compared to the winter (Table 2).

Table 2 Number of follicles visualized at OPU and of recovered oocytes and their quality in summer and winter

Oocyte transcriptome analysis

After quality control, data from 8 libraries (n = 4 for summer and n = 4 for winter) were used for the bioinformatic analyses. The eight cDNA libraries had an approximate average of 53 million clean reads per library, with an average of 98.4% of the reads mapped to the bovine reference genome. The principal component analysis (PCA) in Fig. 4A and hierarchical heatmap in Fig. 4B displayed clear separation and distribution based on the expression of genes for each group. A total of 12,489 and 13,129 genes were expressed (zFPKM > -3 in the four replicates) in the summer and winter groups, respectively with 12,239 genes being mutually expressed during both seasons. Alternatively, 250 genes were expressed exclusively in the summer, while 890 genes were expressed exclusively in the winter (Fig. 4C). Among the top 20 expressed genes, 16 were detected in both groups, including Ubiquitin B (UBB), Serglycin (SRGN), Inhibin Subunit Alpha (INHA), Cytochrome C Oxidase I (COX1), Cytochrome C Oxidase III (COX3), and Heat Shock Protein Family B (Small) Member 1 (HSPB1) (Table 3).

Fig. 4
figure 4

Transcriptomic analyses and differentially expressed genes (DEGs) during summer and winter of oocytes in beef cows. Principal component analysis (PCA; A) and hierarchical heatmap (B) indicating the expression of genes in oocyte’s during summer and winter. Venn diagram (C) showing exclusively and mutually expressed genes between summer and winter and Volcano plot (D) indicating the differentially expressed genes from summer compared to the winter [Individual red and green dots represent the number of up- and downregulated genes, respectively (FC ≥ 2; FDR p-value ≤ 0.05)]. Summer = SUM (n = 4) and Winter = WIN (n = 4)

Table 3 The top 20 most abundant expressed genes in the oocytes from the summer (SUM) and the winter (WIN), represented as the mean of TPM. Common genes in SUM and WIN are bolded

Differential expression analysis indicated a total of 1,386 genes differentially expressed between the two groups (FC ≥ 2; FDR = ≤ 0.05; zFPKM>-3) in all biological replicates of the enriched group (Supp Table 2). During the summer, there was an up-regulation of 446 genes and a downregulation of 940 genes compared to the winter (Fig. 4D). Matrix Metallopeptidase 2 (MMP2) was the top downregulated gene (FC = -1628.99) and Tumor necrosis factor Receptor Superfamily Member 13B (TNFRSF13B) was among the top upregulated genes; (FC = 213.56; Table 4).

Table 4 Top 10 up- and downregulated, genes in the oocytes from the summer (SUM) compared to the winter (WIN) groups

Gene enrichment analysis of the oocytes identified protein digestion and absorption, ABC transporters, oocyte meiosis, and progesterone-mediated oocyte maturation as the top significant pathways upregulated in summer (Fig. 5A). While ECM receptor interaction, phosphoinositide 3-kinase (PI3K)–AKT (PI3K-AKT) signaling, and focal adhesion pathways were the top-significant pathways downregulated (Fig. 5B). Moreover, the meiotic cycle and DNA methylation involved in gamete generation were the top significant biological processes upregulated in summer and extracellular matrix organization was the top downregulated (Fig. 5C & D).

Fig. 5
figure 5

Top 20 enriched pathways (A. Upregulated pathways; B. Downregulated pathways) and biological processes (BP; C. Upregulated BP; D. Downregulated BP) in oocytes collected during summer (SUM) and winter (WIN) in beef cows

Granulosa cells transcriptome analysis

Data from 8 libraries (n = 4 for summer and n = 4 for winter) were used in the bioinformatic analyses following quality control. The eight cDNA libraries had an approximate average of 42 million clean reads per library, with an average of 98.57% of the reads mapped to the bovine reference genome. The PCA (Fig. 6A) and hierarchical heatmap (Fig. 6B) displayed clear separation and distribution based on the expression of genes for each group. A total of 12,504 and 12,030 genes were expressed (zFPKM > -3 in the four replicates) in the summer and winter groups, respectively, with 11,488 genes being mutually expressed in both groups. Alternatively, 1016 genes were expressed exclusively in the summer, while 542 genes were expressed exclusively in the winter (Fig. 6C). Among the top 20 expressed genes, 16 genes were also mutually represented amongst both groups (Table 5), including COX1-3, ND1-6, ATP6, and ATP8 with COX1 and COX3 represented as the top 2 genes in both seasons.

Fig. 6
figure 6

Transcriptomic analyses and differentially expressed genes (DEGs) during summer and winter of granulosa cells in beef cows. Principal component analysis (PCA; A) and hierarchical heatmap (B) indicating the expression of genes in granulosa cells during summer and winter. Venn diagram (C) showing exclusively and mutually expressed genes between summer and winter and Volcano plot (D) indicating the differentially expressed genes from summer compared to the winter [Individual red and green dots represent the number of up- and downregulated genes, respectively (FC ≥ 2; FDR p-value ≤ 0.05)]. Summer = SUM (n = 4) and Winter = WIN (n = 4)

Table 5 The top 20 most abundantly expressed genes in the granulosa cells of the summer (SUM) and the winter (WIN) groups, represented as the mean of TPM. Common genes in SUM and WIN are bolded

Differential expression analysis indicated a total of 2,209 genes significantly differentially expressed between the two seasons (FC ≥ 2; FDR = ≤ 0.05; zFPKM>-3) in all replicates of the enriched group; Supp Table 3). Comparing the granulosa cells collected during the summer and the winter groups, there was an up-regulation of 1083 genes and a down-regulation of 1126 genes (Fig. 6D). Table 6 represents the top 10 up- and downregulated DEGs. Gene enrichment analysis of DEGs in granulosa cell samples identified protein processing in the endoplasmic reticulum and cell cycle as the top-significant upregulated pathways in summer, while chemokine signaling and Th17 cell differentiation were the top downregulated (Fig. 7A & B). When analyzing biological processes enrichment, microtubule-based movement and cell division were the top upregulated, while the inflammatory response was the top downregulated in summer (Fig. 7C & D).

Table 6 Top 10 up- and downregulated genes in the granulosa cells from the summer (SUM) compared to the winter (WIN)
Fig. 7
figure 7

Top 20 enriched pathways (A. Upregulated pathways; B. Downregulated pathways) and biological processes (BP; C. Upregulated BP; D. Downregulated BP) in granulosa cells collected during summer (SUM) and winter (WIN) in beef cows

Comparative gene expression analysis of oocyte and granulosa cells

Differential expression analysis revealed a set of 270 genes commonly dysregulated in both oocyte and granulosa cell datasets (Fig. 8). Within this set, 127 genes exhibited a similar expression pattern in which 13 were upregulated and 114 were downregulated in both oocytes and granulosa cells during summer compared to winter conditions. Conversely, 101 genes were upregulated in granulosa cells and downregulated in oocytes while 42 genes showed the opposite pattern, being upregulated in oocytes, and downregulated in granulosa cells (Fig. 8). Pathway analysis of these 270 common DEGs identified several pathways including transforming growth factor-β (TGF-beta), TNF, mitogen-activated protein kinase (MAPK), and PI3K-Akt signaling pathways, as well as cytokine-cytokine receptor interaction, Th1 and Th2 cell differentiation, focal adhesion, ECM-receptor interaction, and protein digestion and absorption pathways as the top affected pathways. The interaction network of these pathways and their corresponding DEGs is presented in Fig. 9. Most of these pathways involved genes that were mutually downregulated in both datasets, as depicted in Fig. 10.

Fig. 8
figure 8

Commonly and exclusively differentially expressed genes from summer compared to winter collected oocytes (OO) and granulosa cells (GCs)

Fig. 9
figure 9

Top pathways and the related commonly DEGs in the oocytes and granulosa cells in summer compared to winter groups

Fig. 10
figure 10

Top pathways and the related DEGs which exhibited a downregulation in oocytes and granulosa cells in summer compared to winter groups

Discussion

Our study aimed to identify differences in the transcriptome of oocytes and granulosa cells collected from beef cows during the summer and winter months. Throughout our study, the THI during the summer period continually deviated from normal levels, likely indicating that the animals within the study were exposed to environmental heat stress as evidenced by their increased rectal temperatures. In Bos taurus beef cattle, a THI over 75 signifies a level of heat stress [22], which was replicated in the present study with the average summer THI being 79.2. While THI has been commonly used as an indicator of heat stress in livestock [23, 24], it does come with certain limitations given that other factors, such as solar radiation and wind speed, are not included in its calculation [25, 26]. In our study, plasma cortisol concentrations did not differ between collections during the summer and winter. Cortisol is a steroid hormone regulated by the hypothalamic-pituitary-adrenal axis [27], in which previous studies in dairy cattle [28,29,30] have found a significant increase in plasma cortisol concentrations in animals exposed to heat stress. Such discrepancies in cortisol results might be due to the fact that our observations included chronic (long-term) rather than acute (short-term) exposure to levels of heat stress. Additionally, in our study the transcript abundance of HSP70 in leukocytes tended to increase in the summer (P = 0.06), while levels of HSP90 remained stable during both seasons. In peripheral blood mononuclear cells of beef calves, Kim et al. [31] found that HSP70 expression was higher during conditions of heat stress, while the expression of HSP90 showed no differences, concluding that HSP70 exhibits a more sensitive mechanism than HSP90, aiding in the adaptation to heat stress. A major function of HSPs are via their role as indicators of thermotolerance [32], defined as an animal’s ability to balance heat production/dissipation under ambient temperatures above the thermoneutral zone [33]. Therefore, the combination of increased rectal temperatures and the tendency of increased HSP70 expression in leukocytes during the summer, may indicate that our experimental animals have experienced heat stress prior to the summer OPU collections.

Heat stress affects oocyte quality and subsequently oocyte competence [34], nuclear maturation rate [35], fertilization rate [35], and blastocyst production in vitro [34]. Similarly, in the present study while a higher proportion of grade 1 oocytes was collected during winter, the proportion of grade 4 oocytes was higher in summer collections. In addition, heat stress disturbs the synthesis of steroid hormones (i.e., decrease estradiol although increase progesterone concentration in the follicular fluid) [36] which are directly involved in the mechanisms regulating oocyte maturation. The consistency in these results among different studies should be a concern for the beef industry, where there is low adoption of reproductive biotechnologies that would help improve fertility and pregnancy rates in the summer (such as in vitro embryo production and embryo transfer programs) and where reproductive programs consisting of natural mating with no controlled breeding season (i.e., bulls are allowed to breed cows continuously during the year) are predominantly used [37].

In the current study we showed that oocytes and granulosa cells respond to environmental fluctuations between the summer and winter seasons through the activation or inhibition of genes involved in several pathways associated with ovarian function. We reported here that 1,386 genes were differently expressed in oocytes during the summer and winter. The top upregulated protein-coding genes for oocytes collected during the summer were TNFRSF13B, Gastrin (GAST), Dopamine Receptor D5 (DRD5), Pancreatic Polypeptide Receptor 1 (PPYR1), Ephrin A3 (EFNA3), Visual System Homeobox 1 (VSX1), and T Cell Leukemia Homeobox 3 (TLX3). On the other hand, Collagen Type III Alpha 1 Chain (COL3A1), TEA Domain Transcription Factor 4 (TEAD4), TNF Receptor Superfamily Member 8 (TNFRSF8), TNF Superfamily Member 4 (TNFSF4), C1q and TNF Related 5 (C1QTNF5), X-C Motif Chemokine Ligand 1 (XCL1), Epithelial Membrane Protein 1 (EMP1), Annexin A8 Like 1 (ANXA8L1), Transmembrane Protein 26 (TMEM26), and Matrix Metallopeptidase 2 (MMP2) were among the top downregulated genes in summer. These dramatic differences in the transcriptome of oocytes collected during the summer and winter seasons indicate both oocytes response to elevated environmental THI and its subsequent impact on the physiology of the oocyte.

Genes involved in regulating cellular proliferation were upregulated in oocytes collected during the summer season. Among the transcripts upregulated, TNFRSF13B is a tumor necrosis factor receptor superfamily member previously identified in human and mouse oocytes [38]. GAST, which encodes for a peptide hormone key in gastric acid secretion in stomach G cells, was reported to be associated with positive regulation of cell proliferation [39,40,41]. EFNA3 is a member of the ephrin (EPH) family, a group of molecules known for playing a role in embryonic development, cellular proliferation, migration, and adhesion [42,43,44]. DRD5 is expressed in several types of cancers, and treatment with DRD5 agonists can induce apoptosis and autophagy [45]. These upregulated genes are members of the meiotic cell cycle and cell differentiation pathways, enriched in oocytes collected during the summer season. The upregulation of cellular proliferation markers in immature oocytes from summer compared to winter could be a protective response to the challenging conditions caused by heat stress. However, it can also disrupt the normal developmental process of the oocyte which relies on the balance between proliferation and apoptosis in the COCs, leading to suboptimal maturation or developmental competence [46]. Additionally, the cumulus cells surrounding the oocyte normally undergo a shift from proliferation to differentiation as the oocyte matures. Persistent proliferation markers could suggest incomplete differentiation of these supporting cells [47].

Interestingly, Gendelman and Roth (2012) described an abundance of Proto-Oncogene C-Mos (C-MOS), Growth Differentiation Factor 9 (GDF9), POU Class 5 Homeobox 1 (POU5F1), and Glyceraldehyde-3-Phosphate Dehydrogenase (GAPDH) transcripts as higher in MII-stage oocytes collected in the cold season than in those from the hot season [48]. However, in their study, this seasonal variation was not present in GV-stage oocytes [48]. In contrast, our study found that MOS and GDF9 were upregulated in oocytes during the summer season (GV-stage). Also, similar to Ferreira et al. [49], we found an upregulation of Fibroblast Growth Factor 16 (FGF16) and GDF9 in oocytes collected during the summer months compared to those in the winter months. This may signify the fact that oocytes respond to thermal stress in stage dependent manner.

Among the genes downregulated in oocytes collected during the summer season, transmembrane protein 26 (TMEM26) and matrix metalloproteinase 2 (MMP2) were suppressed by more than a thousand-fold. The latter, i.e., MMP2, levels in human follicular fluid were found to be a reliable marker for oocyte maturation in in vitro fertilization and intracytoplasmic sperm injection cycles [50]. Moreover, a recent study by Latorraca et al. [51] identified MMP2 to be present at all phases of oocyte growth (common to all oocyte size groups, from < 60 μm to > 120 μm). Therefore, the significant reduction of such transcripts in oocytes collected during the summer season implies the negative impact of thermal stress on oocytes’ developmental competence by suppressing transcripts critical for their growth and development. Collectively, double the number of transcripts were found to be downregulated in oocytes collected during the summer compared to their winter counterparts. This fact evidenced the potential impact of elevated environmental temperatures on oocytes competence is via the suppression of the expression of transcripts important for their growth and maturation. This has also been evidenced in the type of pathways in which the downregulated transcripts participate (Fig. 5B). A significant proportion of downregulated transcripts were found to be involved in PI3K-Akt signaling and focal adhesion pathways. The PI3K-Akt signaling pathway is reported to be a key regulator of many cellular processes associated with cell proliferation, survival, growth, cytoskeletal rearrangement, and metabolism [52], which makes the pathway to be a potential predictor for the developmental competence of oocytes and successful embryo implantation [53]. Similarly, studies in bovine oocytes revealed that follicular granulosa cells’ expression of the PI3K-Akt signaling pathway to be correlated with the developmental competence of oocytes after parthenogenetic activation [54]. Therefore, our results demonstrate that one of the mechanisms by which environmental thermal stress impacts oocyte competence is via the suppression of transcripts involved in various key pathways regulating follicular development, oocyte maturation, and further embryonic developmental potential.

Granulosa cells are known to mediate oocyte developmental process via an active bidirectional communication with the oocyte [55, 56]. Therefore, any significant negative impact of environmental seasonal changes on granulosa cell physiology would have a direct impact on ovarian function and oocyte developmental processes. Transcriptome analysis of granulosa cells form summer and winter seasons revealed that among the 2,209 differentially expressed genes, several members of the Heat Shock proteins family (Heat Shock Protein Family A (Hsp70) Member 5 (HSPA5), Heat Shock Protein 90 Beta Family Member 1 (HSP90B1), DnaJ Heat Shock Protein Family (Hsp40) Member B9 (DNAJB9), B11 (DNAJB11), B14 (DNAJB14), B13 (DNAJB13), and C5 (DNAJC25) were found to be upregulated in granulosa cells during the summer season as a response to higher environmental temperatures. These genes have been previously described as components of the Heat Shock Protein cascade in response to heat stress [57, 58], and have been shown to be elevated in granulosa cells after in vitro exposure to acute heat stress [59, 60]. In addition, here we report that several members of the axonemal dynein complex members including Dynein Axonemal Heavy Chains 1 (DNAH1), 5 (DNAH5), 6 (DNAH6), 7 (DNAH7), 9 (DNAH9), 10 (DNAH10), 11 (DNAH11), and 12 (DNAH12) to be upregulated in the summer months. Moreover, DNAH6, DNAH10, and DNAH11 were in the top 10 upregulated genes in the granulosa cells during summer. Dyneins are a family of cytoskeletal motor proteins that form the microtubules in cells. Dyneins have been localized in follicles (granulosa cells and oocytes) [61, 62], and in mice, the inhibition of dynein proteins significantly increased the number of growing follicles [63].

Other class of transcripts related to Zona Pellucida Glycoprotein 2 (ZP2), a critical component of the Zona Pellucida, was in the top 10 upregulated genes in the granulosa cells during summer. Similar studies have also previously indicated that heat stress impacts the ZP function and therefore disrupts the anti-polyspermy system of the oocyte, which resulted in increasing polyspermy [64] and increased number of pores and a more significant percentage of oocytes with amorphous ZPs [65] as a result of thermal stress. Collectively, our data and also from others suggest that in vivo, granulosa cells are also susceptible to seasonal heat stress provoking changes in cell transcriptome dynamics, which could be possibly related to the decrease in oocyte quality in the summer.

As observed in oocytes, granulosa cells collected during summer showed upregulation of genes involved in protein processing in endoplasmic reticulum, while downregulated genes are involved in chemokine signaling pathways. Accumulation of unfolded or misfolded proteins in the endoplasmic reticulum due to various environmental stressors induced the onset of ER stress, which ultimately leads to cellular apoptosis [66]. One of the coordinated responses to accumulated unfolded protein in the ER is called unfolded protein response (UPR). Therefore, the upregulation of genes associated with protein processing in ER in the current study revealed that increased protein processing in the ER is one of the mechanisms in which granulosa cells respond to elevated environmental temperature.

Integration of the oocyte and granulosa cells transcriptome data revealed differential regulation of 127 genes in common with a similar direction of expression in both oocytes and granulosa cells in response to environmental temperature. The majority of the transcripts (114 genes) were found to be suppressed in both oocytes and granulosa cells collected during the summer season compared to the winter. Pathway analysis for these commonly suppressed transcripts due to thermal stress in oocytes and granulosa cells revealed their enrichment in several pathways associated with ovarian function, follicle and oocyte development (Fig. 10). The involvement of the TGF-beta and PI3K-Akt pathways suggests alterations in cell growth, apoptosis, and metabolic processes, which are critical for follicular maturation and oocyte quality [53, 67]. Additionally, changes in focal adhesion and ECM-receptor interactions indicate potential disruptions in oocyte-follicle communication and intracellular signaling [68] due to the elevated summer temperature. Investigating the functional consequences of these gene expression changes on further embryonic development in vivo could provide deeper insights into how environmental stressors affect reproductive efficiency. This is the first evidence indicating how follicular cells (granulosa cells) and gametes (oocytes) are affected by or respond to environmental elevated temperature in the ovarian follicle.

Conclusion

Our results indicate that cows exposed to seasonal fluctuations of heat stress dramatically alter their granulosa cells and oocytes transcriptome, which may in part be responsible for negatively impacting follicular physiology. Specified genes and their associated pathways and biological processes pose an essential function in bovine granulosa cells and oocytes’ response to heat stress, potentially serving as mechanisms for future targets to mitigate the impact of heat stress within the intrafollicular environment.

Data availability

The raw FASTQ files and processed CSV files of the RNA sequencing data have been deposited in the NCBIs Gene Expression Omnibus (GEO) repository with the accession numbers GSE235170 and GSE235171.

Abbreviations

ANXA8L1:

Annexin A8 Like 1

ATP6:

Mitochondrially Encoded ATP Synthase Membrane Subunit 6

ATP8:

Mitochondrially encoded ATP synthase membrane subunit 8

BP:

Biological processes

C1QTNF5:

C1q and TNF Related 5

C-MOS:

Proto-Oncogene C-Mos

COCs:

Cumulus-oocyte-complexes

COL3A1:

Collagen Type III Alpha 1 Chain

COX1:

Cytochrome C Oxidase I

COX3:

Cytochrome C Oxidase III

CV:

Coefficient of variation

DEGs:

Differentially expressed genes

DNAH 1, 1, 5, 6, 7, 9, 10, 11, and 12:

Dynein Axonemal Heavy Chains 1, 5, 6, 7, 9, 10, 11, and 12

DNAJB 11, 13, 14, and 90:

DnaJ Heat Shock Protein Family (Hsp40) Members B11 B13, B14, and B90

DNAJC25:

DnaJ Heat Shock Protein Family (Hsp40) Member C25

DRD5:

Dopamine Receptor D5

ECM:

Extracellular Matrix

EFNA3:

Ephrin A3

EMP1:

Epithelial Membrane Protein 1

EVs:

Extracellular vesicles

FAWN:

Florida Automated Weather Network

FDR:

False discovery rate

FGF16:

Fibroblast Growth Factor 16

GAPDH:

Glyceraldehyde-3-Phosphate Dehydrogenase

GAST:

Gastrin

GDF9:

Growth Differentiation Factor 9

GEO:

NCBI’s Gene Expression Omnibus

GLM:

Generalized Linear Model

GV:

Germinal vesicle

Hsp40:

DnaJ Heat Shock Protein Family

HSP70:

Heat shock protein 70

HSP90:

Heat shock protein 90

HSP90B1:

Heat Shock Protein 90 Beta Family Member 1

HSPA5:

Heat Shock Protein Family A (Hsp70) Member 5

HSPB1:

Heat Shock Protein Family B (Small) Member 1

INHA:

Inhibin Subunit Alpha

MAPK:

Mitogen-activated protein kinase

MMP2:

Matrix Metallopeptidase 2

ND1-6:

NADH dehydrogenase subunits 1–6

OPU:

Ovum Pick up

PCA:

Principal component analysis

PI3K-AKT:

Phosphoinositide 3-kinase (PI3K)–AKT pathway

POU5F1:

POU Class 5 Homeobox 1

PPYR1:

Pancreatic Polypeptide Receptor 1

qPCR:

Quantitative real-time PCR

SRGN:

Serglycin

TEAD4:

TEA Domain Transcription Factor 4

TGF-beta:

Transforming growth factor-β

THI:

Temperature humidity index

TLX3:

T Cell Leukemia Homeobox 3

TMEM26:

Transmembrane Protein 26

TMM normalization:

Trimmed mean of the M-values normalization method

TNF:

Tumor necrosis factor

TNFRSF13B:

TNF Receptor Superfamily Member 13B

TNFRSF8:

TNF Receptor Superfamily Member 8

TNFSF4:

TNF Superfamily Member 4

TPM:

Transcripts per million

UBB:

Ubiquitin B

UPR:

Unformed protein response

VSX1:

Visual System Homeobox 1

XCL1:

X-C Motif Chemokine Ligand 1

ZP2:

Zona Pellucida Glycoprotein 2

References

  1. Thornton P, Nelson G, Mayberry D, Herrero M. 2022. Impacts of heat stress on global cattle production during the 21st century: a modelling study. Lancet Planet Health. 2022;6:e192–e201. https://doi.org/10.1016/S2542-5196(22)00002-X

  2. Fernandez-Novo A, Pérez-Garnelo SS, Villagrá A, Pérez-Villalobos N, Astiz S. The effect of stress on Reproduction and Reproductive Technologies in Beef Cattle-A review. Animals. 2020;10:2096–119. https://doi.org/10.3390/ani10112096.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Badinga L, Thatcher WW, Diaz T, Drost M, Wolfenson D. Effect of environmental heat stress on follicular development and steroidogenesis in lactating Holstein cows. Theriogenology. 1993;39:797–810. https://doi.org/10.1016/0093-691x(93)90419-6.

    Article  CAS  PubMed  Google Scholar 

  4. Khan A, Khan MZ, Umer S, Khan IM, Xu H, Zhu H, Wang Y. Cellular and molecular adaptation of bovine granulosa cells and oocytes under heat stress. Animals. 2020b;10:110. https://doi.org/10.3390/ani10010110.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Roth Z. Effect of heat stress on Reproduction in dairy cows: insights into the Cellular and molecular responses of the oocyte. Annu Rev Anim Biosci. 2017;5:151–70. https://doi.org/10.1146/annurev-animal-022516-022849.

    Article  CAS  PubMed  Google Scholar 

  6. Rocha A, Randel RD, Broussard JR, Lim JM, Blair RM, Roussel JD, Hansel W. High environmental temperature and humidity decrease oocyte quality in Bos taurus but not in Bos indicus cows. Theriogenology. 1998;49:657–65. https://doi.org/10.1016/s0093-691x(98)00016-8.

    Article  CAS  PubMed  Google Scholar 

  7. Payton RR, Rispoli LA, Nagle KA, Gondro C, Saxton AM, Voy BH, Edwards JL. Mitochondrial-related consequences of heat stress exposure during bovine oocyte maturation persist in early embryo development. J Reprod Dev. 2018;64:243–51. https://doi.org/10.1262/jrd.2017-160.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Li J, Gao H, Tian Z, Wu Y, Wang Y, Fang Y, Lin L, Han Y, Wu S, Haq IU, Zang S. Effects of chronic heat stress on granulosa cell apoptosis and follicular atresia in mouse ovary. J Anim Sci Biotechnol. 2016;7:57. https://doi.org/10.1186/s40104-016-0116-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Gad A, Joyce K, Menjivar NG, Heredia D, Rojas CS, Tesfaye D, Gonella-Diaza A. Extracellular vesicle-microRNAs mediated response of bovine ovaries to seasonal environmental changes. J Ovarian Res. 2023;16:101. https://doi.org/10.1186/s13048-023-01181-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. International Embryo Technology Society. https://www.iets.org. Accessed 13 March 2023.

  11. Florida Automated Weather Network (FAWN). https://fawn.ifas.ufl.edu/data/reports/. Accessed 10 January 2022.

  12. NRC. A guide to environmental research on animals - National Research Council (US). Committee on Physiological Effects of Environmental Factors on Animals. Natl. Acad. Sci., Washington,DC; 1971.

  13. Xie F, Xiao P, Chen D, Xu L, Zhang B. miRDeepFinder: a miRNA analysis tool for deep sequencing of plant small RNAs. Plant Mol Biol. 2012;80:75–84. https://doi.org/10.1007/s11103-012-9885-2.

    Article  CAS  Google Scholar 

  14. Xie F, Wang J, Zhang B. RefFinder: a web-based tool for comprehensively analyzing and identifying reference genes. Funct Integr Genomics. 2023;23:125. https://doi.org/10.1007/s10142-023-01055-7.

    Article  CAS  PubMed  Google Scholar 

  15. Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 2001;29:e45. https://doi.org/10.1093/nar/29.9.e45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Robinson MD, Oshlack A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 2010;11:R25. https://doi.org/10.1186/gb-2010-11-3-r25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hart T, Komori HK, LaMere S, Podshivalova K, Salomon DR. Finding the active genes in deep RNA-seq gene expression studies. BMC Genomics. 2013;14:778. https://doi.org/10.1186/1471-2164-14-778.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. Royal Stati Soci: Ser B. 1995;57:289–300. https://doi.org/10.1111/j.2517-6161.1995.tb02031.x.

    Article  Google Scholar 

  19. Database. for Annotation, Visualization, and Integrated Discovery (DAVID) Bioinformatics. http://david.abcc.ncifcrf.gov/. Accesssed 23 May 2022.

  20. Ogata H, Goto S, Sato K, Fujibuchi W, Bono H, Kanehisa M. KEGG: Kyoto Encyclopedia of genes and genomes. Nucleic Acids Res. 1999;27:29–34. https://doi.org/10.1093/nar/27.1.29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. LCI. Patterns of transit losses. Livestock Conservation, Inc. Omaha, NE; 1970.

  22. St-Pierre NR, Cobanov B, Schnitkey G. Economic losses from heat stress by US livestock industries. J Dairy Sci. 2003;86:E52–77. https://doi.org/10.3168/jds.S0022-0302(03)74040-5.

    Article  Google Scholar 

  23. Ingraham RH, Gillette DD, Wagner WD. Relationship of temperature and humidity to conception rate of Holstein cows in subtropical climate. J Dairy Sci. 1974;57:476–81. https://doi.org/10.3168/jds.S0022-0302(74)84917-9.

    Article  CAS  PubMed  Google Scholar 

  24. Dikmen S, Hansen PJ. Is the temperature-humidity index the best indicator of heat stress in lactating dairy cows in a subtropical environment? J Dairy Sci. 2009;92:109–16. https://doi.org/10.3168/jds.2008-1370.

    Article  CAS  PubMed  Google Scholar 

  25. Gaughan JB, Mader TL, Holt SM, Lisle A. A new heat load index for feedlot cattle. J Anim Sci. 2008;86:226–34. https://doi.org/10.2527/jas.2007-0305.

    Article  CAS  PubMed  Google Scholar 

  26. Brown-Brandl TM. Understanding heat stress in beef cattle. R Bras Zootec. 2018;47:e20160414. https://doi.org/10.1590/rbz4720160414.

    Article  Google Scholar 

  27. Brown EJ, Vosloo A. The involvement of the hypothalamopituitary-adrenocortical axis in stress physiology and its significance in the assessment of animal welfare in cattle. Onderstepoort J Vet Res. 2017;84:e1–9. https://doi.org/10.4102/ojvr.v84i1.1398.

    Article  CAS  PubMed  Google Scholar 

  28. Christison GI, Johnson HD. Cortisol turnover in heat-stressed cows. J Anim Sci. 1972;35:1005–10. https://doi.org/10.2527/jas1972.3551005x.

    Article  CAS  PubMed  Google Scholar 

  29. Abilay TA, Mitra R, Johnson HD. Plasma cortisol and total progestin levels in Holstein steers during acute exposure to high environmental temperature (42 C) conditions. J Anim Sci. 1975;41:113–7. https://doi.org/10.2527/jas1975.411113x.

    Article  CAS  PubMed  Google Scholar 

  30. Chen S, Wang J, Peng D, Li G, Chen J, Gu X. Exposure to heat-stress environment affects the physiology, circulation levels of cytokines, and microbiome in dairy cows. Sci Rep. 2018. https://doi.org/10.1038/s41598-018-32886-1.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Kim WS, Ghassemi Nejad J, Peng DQ, Jung US, Kim MJ, Jo YH, Jo JH, Lee JS, Lee HG. Heat-shock proteins gene expression in peripheral blood mononuclear cells as an indicator of heat stress in beef calves. Animals. 2020;10:895. https://doi.org/10.3390/ani10050895.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Lindquist S. The heat-shock response. Annu Rev Biochem. 1986;55:1151–91. https://doi.org/10.1146/annurev.bi.55.070186.005443.

    Article  CAS  PubMed  Google Scholar 

  33. Hassan FU, Nawaz A, Rehman MS, Ali MA, Dilshad SMR, Yang C. Prospects of HSP70 as a genetic marker for thermo-tolerance and immuno-modulation in animals under climate change scenario. Anim Nutr. 2019;5:340–50. https://doi.org/10.1016/j.aninu.2019.06.005.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Al-Katanani YM, Paula-Lopes FF, Hansen PJ. Effect of season and exposure to heat stress on oocyte competence in Holstein cows. J Dairy Sci. 2002;85:390–6. https://doi.org/10.3168/jds.s0022-0302(02)74086-1.

    Article  CAS  PubMed  Google Scholar 

  35. Lenz RW, Ball GD, Leibfried ML, Ax RL, First NL. In vitro maturation and fertilization of bovine oocytes are temperature-dependent processes. Biol Reprod. 1983;29:173–9. https://doi.org/10.1095/biolreprod29.1.173.

    Article  CAS  PubMed  Google Scholar 

  36. Roth Z, Meidan R, Shaham-Albalancy A, Braw-Tal R, Wolfenson D. Delayed effect of heat stress on steroid production in medium-sized and preovulatory bovine follicles. Reproduction. 2001;121:745–51. https://doi.org/10.1530/rep.0.1210745.

    Article  CAS  PubMed  Google Scholar 

  37. NAHMS. Beef 2017: Beef Cow-Calf Management Practices in the United States. https://www.aphis.usda.gov/animal_health/nahms/beefcowcalf/downloads/beef2017/Beef2017_dr_PartI.pdf (2017). Accessed 1 Nov 2021.

  38. Haouzi D, Boumela I, Chebli K, Hamamah S. Global, Survival, and apoptotic transcriptome during mouse and human early embryonic development. Biomed Res Int. 2018;2018:5895628. https://doi.org/10.1155/2018/5895628.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. De Hauwer C, Camby I, Darro F, Migeotte I, Decaestecker C, Verbeek C, Danguy A, Pasteels JL, Brotchi J, Salmon I, Van Ham P. Gastrin inhibits motility, decreases cell death levels, and increases proliferation in human glioblastoma cell lines. J Neurobiol. 1998;37:373–82. https://doi.org/10.1002/(sici)1097-4695(19981115)37:3%3C373::aid-neu3%3E3.3.co;2-8.

    Article  PubMed  Google Scholar 

  40. Dockray G, Dimaline R, Varro A. Gastrin: old hormone, new functions. Pflügers Archiv. 2005;449:344–55. https://doi.org/10.1007/s00424-004-1347-5.

    Article  CAS  PubMed  Google Scholar 

  41. Meng S, Shi C, Jia Y, Fu M, Zhang T, Wu N, Han H, Shen H. A combined clinical and specific genes’ model to predict live birth for in vitro fertilization and embryo transfer patients. BMC Pregnancy Childbirth. 2023;23:702. https://doi.org/10.1186/s12884-023-05988-6.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Frisén J, Holmberg J, Barbacid M. Ephrins and their eph receptors: multitalented directors of embryonic development. EMBO J. 1999;18:5159–65. https://doi.org/10.1093/emboj/18.19.5159.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Vihanto MM, Plock J, Erni D, Frey BM, Frey FJ, Huynh-Do U. Hypoxia upregulates expression of eph receptors and ephrins in mouse skin. FASEB J. 2005;19:1689–91. https://doi.org/10.1096/fj.04-3647fje.

    Article  CAS  PubMed  Google Scholar 

  44. Buensuceso AV, Deroo BJ. The Ephrin Signaling Pathway regulates morphology and adhesion of mouse granulosa cells in Vitro. Biol Reprod. 2013;88:1–12. https://doi.org/10.1095/biolreprod.112.100123.

    Article  CAS  Google Scholar 

  45. Wang X, Wang ZB, Luo C, Mao XY, Li X, Yin JY, Zhang W, Zhou HH, Liu ZQ. The prospective value of dopamine receptors on bio-behavior of tumor. J Cancer. 2019;10:1622–32. https://doi.org/10.7150/jca.27780.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Chermuła B, Brązert M, Jeseta M, Ożegowska K, Sujka-Kordowska P, Konwerska A, Bryja A, Kranc W, Jankowski M, Nawrocki MJ, Kocherova I, Celichowski P, Borowiec B, Popis M, Budna-Tukan J, Antosik P, Bukowska D, Brussow KP, Pawelczyk L, Bruska M, Zabel M, Nowicki M, Kempisty B. The Unique mechanisms of Cellular Proliferation, Migration and apoptosis are regulated through Oocyte Maturational Development-A Complete Transcriptomic and histochemical study. Int J Mol Sci. 2018;20(1):84. https://doi.org/10.3390/ijms20010084. PMID: 30587792; PMCID: PMC6337548.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Turathum B, Gao EM, Chian RC. The function of Cumulus cells in Oocyte Growth and Maturation and in subsequent ovulation and fertilization. Cells. 2021;10(9):2292. https://doi.org/10.3390/cells10092292. PMID: 34571941; PMCID: PMC8470117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gendelman M, Roth Z. In vivo vs. in vitro models for studying the effects of elevated temperature on the GV-stage oocyte, subsequent developmental competence and gene expression. Anim Reprod Sci. 2012;134:125–34. https://doi.org/10.1016/j.anireprosci.2012.07.009.

    Article  CAS  PubMed  Google Scholar 

  49. Ferreira RM, Chiaratti MR, Macabelli CH, Rodrigues CA, Ferraz ML, Watanabe YF, Smith LC, Meirelles FV, Baruselli PS. The infertility of repeat-breeder cows during summer is associated with decreased mitochondrial DNA and increased expression of mitochondrial and apoptotic genes in oocytes. Biol Reprod. 2016;94(3):66–1.

    Article  PubMed  Google Scholar 

  50. Yang WJ, Liu FC, Hsieh JS, Chen CH, Hsiao SY, Lin CS. Matrix metalloproteinase 2 level in human follicular fluid is a reliable marker of human oocyte maturation in in vitro fertilization and intracytoplasmic sperm injection cycles. Reprod Biol Endocrinol. 2015;13:102. https://doi.org/10.1186/s12958-015-0099-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Latorraca LB, Galvão A, Rabaglino MB, D’Augero JM, Kelsey G, Fair T. Single-cell profiling reveals transcriptome dynamics during bovine oocyte growth. BMC Genomics. 2024;25:335. https://doi.org/10.1186/s12864-024-10234-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Makker A, Mati Goel M, Mahdi AA. PI3K/PTEN/Akt and TSC/mTOR signaling pathways, ovarian dysfunction, and infertility: an update. J Mol Endocrinol. 2014;53(3):R102–18.

    Article  Google Scholar 

  53. Kalous J, Aleshkina D, Anger M. A role of PI3K/Akt signaling in oocyte maturation and early embryo development. Cells. 2023;12(14):1830.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Andrade GM, Da Silveira JC, Perrini C, Del Collado M, Gebremedhn S, Tesfaye D, Meirelles FV, Perecin F. The role of the PI3K-Akt signaling pathway in the developmental competence of bovine oocytes. PLoS ONE. 2017;12(9):e0185045.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Eppig JJ. Intercommunication between mammalian oocytes and companion somatic cells. BioEssays. 1991;13:569–74. https://doi.org/10.1002/bies.950131105.

    Article  CAS  PubMed  Google Scholar 

  56. Kidder GM, Vanderhyden BC. Bidirectional communication between oocytes and follicle cells: ensuring oocyte developmental competence. Can J Physiol Pharmacol. 2010;88:399–413. https://doi.org/10.1139/y10-009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Kampinga HH, Hageman J, Vos MJ, Kubota H, Tanguay RM, Bruford EA, Cheetham ME, Chen B, Hightower LE. Guidelines for the nomenclature of the human heat shock proteins. Cell Stress Chaperones. 2009;14:105–11. https://doi.org/10.1007/s12192-008-0068-7.

    Article  CAS  PubMed  Google Scholar 

  58. Hu C, Yang J, Qi Z, Wu H, Wang B, Zou F, Mei H, Liu J, Wang W, Liu Q. Heat shock proteins: Biological functions, pathological roles, and therapeutic opportunities. MedComm. 2022;3:e161. https://doi.org/10.1002/mco2.161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Khan A, Dou J, Wang Y, Jiang X, Khan MZ, Luo H, Usman T, Zhu H. Evaluation of heat stress effects on cellular and transcriptional adaptation of bovine granulosa cells. J Anim Sci Biotechnol. 2020a;11:25. https://doi.org/10.1186/s40104-019-0408-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Sammad A, Luo H, Hu L, Zhu H, Wang Y. Transcriptome reveals granulosa cells coping through redox, inflammatory and metabolic mechanisms under acute heat stress. Cells. 2022;11:1443. https://doi.org/10.3390/cells11091443.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Li MG, McGrail M, Serr M, Hays TS. Drosophila cytoplasmic dynein, a microtubule motor that is asymmetrically localized in the oocyte. J Cell Biol. 1994;126:1475–94. https://doi.org/10.1083/jcb.126.6.1475.

    Article  CAS  PubMed  Google Scholar 

  62. Kehoe S, Jewgenow K, Johnston PR, Mbedi S, Braun BC. Signalling pathways and mechanistic cues highlighted by transcriptomic analysis of primordial, primary, and secondary ovarian follicles in domestic cat. Sci Rep. 2021;11:2683. https://doi.org/10.1038/s41598-021-82051-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Nagamatsu G, Shimamoto S, Hamazaki N, Nishimura Y, Hayashi K. Mechanical stress accompanied with nuclear rotation is involved in the dormant state of mouse oocytes. Sci Adv. 2019;5:eaav9960. https://doi.org/10.1126/sciadv.aav9960.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Sakatani M, Yamanaka K, Balboula AZ, Takenouchi N, Takahashi M. Heat stress during in vitro fertilization decreases fertilization success by disrupting anti-polyspermy systems of the oocytes. Mol Reprod Dev. 2015;82:36–47. https://doi.org/10.1002/mrd.22441.

    Article  CAS  PubMed  Google Scholar 

  65. Báez F, Camargo Á, Reyes AL, Márquez A, Paula-Lopes F, Viñoles C. Time-dependent effects of heat shock on the zona pellucida ultrastructure and in vitro developmental competence of bovine oocytes. Reprod Biol. 2019;19(2):195–203.

    Article  PubMed  Google Scholar 

  66. Tabas I, Ron D. Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress. Nat Cell Biol. 2011;13(3):184–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Patton BK, Madadi S, Pangas SA. Control of ovarian follicle development by TGF-β family signaling. Curr Opin Endocr Metabolic Res. 2021;18:102–10.

    Article  CAS  Google Scholar 

  68. McGinnis LK, Kinsey WH. Role of focal adhesion kinase in oocyte-follicle communication. Mol Reprod Dev. 2015;82(2):90–102.

    Article  CAS  PubMed  Google Scholar 

  69. Hooper HB, Titto CG, Gonella-Diaza AM, Henrique FL, Pulido-Rodríguez LF, Longo ALS, Leme-Dos-Santos TMDC, Geraldo ACAPM, Pereira AMF, Binelli M, Balieiro JCC, Titto ELA. Heat loss efficiency and HSP gene expression of Nellore cows in tropical climate conditions. Int J Biometeorol. 2019;63:1475–86. https://doi.org/10.1007/s00484-018-1576-5.

    Article  PubMed  Google Scholar 

  70. Bettegowda A, Patel OV, Ireland JJ, Smith G. Quantitative analysis of messenger RNA abundance for ribosomal protein L-15, cyclophilin-A, phosphoglycerokinase, β-glucuronidase, glyceraldehyde 3-phosphate dehydrogenase, β-actin, and histone H2A during bovine oocyte maturation and early embryogenesis. Mol Reprod Dev. 2006;73:267–78. https://doi.org/10.1002/mrd.20333.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Kamryn Joyce was supported by a National Needs Fellowship from USDA (NIFA Grant 2021-38420-34067). The authors thank Jhon Fredy Rodriguez and Edna Ballesteros from Breeding Tech LLC (Madisonville – Texas) for their technical assistance in conducting the OPU procedures. Additionally, the authors thank the NFREC’s Beef Unit crew for their assistance with this experiment.

Funding

Nothing to declare.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: D.T., A.G.D.; Experimental work: K.J., A.G., N.G.M., S.G., D.H., G.D., M.C.L.D., J.B.; Data analysis: K.J., A.G, N.G.M., Interpretation of data: K.J., A.G., N.G.M. A.G.D.; Writing of original draft: K.J., A.G., N.G.M.; Manuscript editing: A.G., N.G.M., D.T., A.G.D.; Supervision: D.T., A.G.D.; Funding acquisition: D.T., A.G.D.; All authors have read and agreed to the published version of this manuscript.

Corresponding author

Correspondence to Angela Gonella-Diaza.

Ethics declarations

Ethics approval and consent to participate

All procedures involving cows were approved by the Animal Care and Use committee of the University of Florida (Approval Number: IACUC202200000706).

Consent for publication

Not applicable.

Clinical trial number

Not applicable.

Footnotes

N/A.

Disclaimer

We declare that in part some of the results described in this manuscript were adapted from Kamryn Joyce’s Master’s Thesis.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Joyce, K., Gad, A., Menjivar, N.G. et al. Seasonal environmental fluctuations alter the transcriptome dynamics of oocytes and granulosa cells in beef cows. J Ovarian Res 17, 201 (2024). https://doi.org/10.1186/s13048-024-01530-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13048-024-01530-0

Keywords