SciELO - Scientific Electronic Library Online

 
vol.12 número4Efecto de la aplicación intrauterina de ozono sobre la migración de neutrófilos y la endometritis subclínica en ganado lecheroDigestibilidad ileal estandarizada de la proteína y aminoácidos de la pasta de ajonjolí en cerdos en crecimiento índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • No hay artículos similaresSimilares en SciELO

Compartir


Revista mexicana de ciencias pecuarias

versión On-line ISSN 2448-6698versión impresa ISSN 2007-1124

Rev. mex. de cienc. pecuarias vol.12 no.4 Mérida oct./dic. 2021  Epub 06-Jun-2022

https://doi.org/10.22319/rmcp.v12i4.5625 

Technical notes

In silico analysis of gene expression in granulosa cells of preovulatory follicles in two species of bovines

Jesús Alfredo Berdugo-Gutiérreza  * 

Ariel Marcel Tarazona-Moralesb 

José Julián Echeverry-Zuluagaa 

Albeiro López-Herreraa 

a Universidad Nacional de Colombia- Sede Medellín. Facultad de Ciencias Agrarias. Grupo de Investigación BIOGEM. Carrera 65 No 59 a 110. Medellín, Colombia.

b Universidad Nacional de Colombia- Sede Medellín. Facultad de Ciencias Agrarias. Grupo de Investigación BIOGENESIS.


Abstract

Buffaloes and cattle are two species of bovines with great similarity in their reproductive physiology, but at the same time with great difference in their reproductive parameters. The objective of this work is to compare gene expression in granulosa cells of preovulatory follicles of these two species, based on information available in the literature, existing transcriptome repositories and functional analysis using Ingenuity Pathway Analysis. Only two independent studies comparing buffalo and cattle in terms of gene expression in granulosa cells of preovulatory follicles were found. Expression data were analyzed independently and in combination. It was found that, between buffaloes and cattle, there is practically no correspondence between the processes evaluated, neither in the canonical pathways, nor in the upstream regulators, only some correspondence is found between the networks and physiological aspects of each process. It is concluded that each species has a different way of carrying out the same process and that each event should be studied according to the needs of the researchers.

Key words Bovines; Granulosa; Transcriptomics; Follicle; In Silico

Resumen

Los búfalos y vacunos son dos especies de bovinos con gran parecido en su fisiología reproductiva, pero a la vez con gran diferencia en sus parámetros reproductivos. El objetivo del presente trabajo es comparar la expresión génica en células de granulosa de folículos preovulatorios de estas dos especies, basados en información disponible en la literatura, los repositorios de transcriptomas existentes y en el análisis funcional usando Ingenuity Pathway Analysis. Sólo se encontraron dos estudios independientes en los que se comparan búfalos y vacunos en cuanto a la expresión génica en células de la granulosa de folículos preovulatorios. Se analizaron los datos de expresión de forma independiente y combinada. Se encontró que, entre búfalos y vacunos, prácticamente no hay correspondencia entre los procesos evaluados, ni en las vías canónicas, ni en los reguladores corriente arriba, solamente se encuentra alguna correspondencia entre las redes y aspectos fisiológicos de cada proceso. Se concluye que cada especie tiene forma diferente de realizar el mismo proceso y que debe investigarse cada evento de acuerdo con las necesidades de los investigadores.

Palabras clave Bovinos; Granulosa; Transcriptómica; Folículo; In Silico

Buffaloes (Bubalus bubalis) and cattle (Bos taurus) are bovines closely related that are only differentiated by their mitochondrial DNA1. Buffaloes and cattle are seasonal polyestrous, with two or three follicular waves per estrous cycle2; however, under the same environmental and management conditions, their reproductive function is different, evidenced by the birth rate, expression of heat and response to reproductive biotechnologies. It has also been described that the buffalo has smaller ovaries3, different follicular diameter at the time of deviation and ovulation4, lower quality of oocytes and lower rate of embryo production compared to cattle. Li et al5 found 40 loci (associated with 28 genes) that could be related to reproductive parameters such as age at first, second and third calving, days open, services per conception and calving interval5,6. The formation of a competent oocyte depends on follicular development, in one the formation of a preantral follicle with an oocyte capable of forming the individual and the second is the development of this follicle until ovulation, associated with all the endocrine changes that must happen in the female to achieve it7,8.

The control of the follicular population depends on the function performed by the anti-Müllerian hormone (AMH), those who are going to ovulate must acquire receptors for follicle-stimulating hormone (FSH), finally some follicle gains receptors for LHr8,which generates a decrease in the speed of growth that is maintained until the preovulatory peak of LH9,10.

It has been reported that the buffalo oocyte is of poor quality when evaluated with the parameters normally used for cattle, and although the reasons are not known in detail, it has been proposed as a cause: an adverse follicular microenvironment, which has motivated researchers to study the proteins of the buffalo follicular fluid11. The existing communication between granulosa cells and the oocyte has been shown to play a critical role in producing good quality oocytes12. Nowadays, it is feasible the in-depth analysis of reproductive events in the female bovine. It is undeniable the progress achieved with microarrays and other systems used in transcriptomics, the data are deposited in repositories, which can be analyzed and allow the researcher to evaluate their premises. However, it is a heterogeneous base due to all the technical aspects and computational tools used for its analysis13. In a recent study, Khan et al10 generated an interactive interface called GranulosaIMAGE, which provides information on gene expression profiles and their isoforms in granulosa cells of cattle in different states of folliculogenesis (http://emb bioinfo.fsaa.ulaval.ca/granulosaIMAGE/).

An important tool is Ingenuity Pathway Analysis (IPA), which is a database used for the analysis, integration and interpretation of data related to experiments based on information obtained by RNA-seq, principally, IPA allows, in a single analysis, identifying regulatory molecules, which facilitates the explanation of expression patterns, predicting the consequences of suggested control on cell biology and on the appearance of diseases14. The program makes the calculations based on algorithms and experimental data15, in which the genes differentially expressed in the experiment are associated with those that have been most frequently reported on a particular function or metabolic pathway16 and its control systems17.

Developments in bioinformatics allow the study of biological phenomena in a complex way: in this case, the differences in the gene expression of buffalo and cattle follicles, in a more global way, which allow at least to consider all the possible genes that are expressed at a given time, showing the details of gene expression and the possible alternatives that nature has generated to carry out the same process. The objective of this work is to evaluate whether there are differences in the gene expression of granulosa cells of buffalo and cattle preovulatory follicles tending to seek explanations of the differential of the response to reproductive biotechnologies observed between the aforementioned species.

This work was carried out in two phases: the first, an exhaustive bibliographic search was carried out in the databases of PUBMED and Google Scholar, focusing on information corresponding to the analysis of gene expression between buffaloes and cows and whose files were deposited in GEO (Gene Expression Omnibus) until March 2018, the second, the analysis of the reports found was carried out. The inclusion criterion was: experiments where there was comparison of data; or if there was no comparison, that the data were obtained in the same way and processed in a similar way.

Two studies of transcriptomes of preovulatory follicle were found18,19. In the case of cows, RNA was extracted with RNeasy mini kit (Qiagen) and in buffalo with TRIZOL followed by purification with RNeasy mini kit, cDNA was obtained in both cases, and they were hybridized with an Affimetrix Chip (Affymetrix Gene - Chip Bovine Genome Arrays), which contained 24,128 probes, representing 23,000 transcripts and their variants, including 19,000 unigene clusters. Those genes with a change in expression ≥2 with a false discovery rate (FDR < 0.05) were considered as expressed differently, subsequently, a semi-quantitative RT-PCR was performed for the validation of the results with the following genes: LH receptor (LHR), progesterone receptor (PR) and cyclooxygenase COX-2 in buffaloes, and two cytochrome P450 genes related to estrogen production, Cytochrome P450 CPY19A1, CPY17A1, 18s, for cattle.

Subsequently, the raw data from each microarray were corrected by background noise removal and normalization (loess) using the FlexArray 1.6.1 software, and all data were exported to excel (Microsoft Office) files to be functionally analyzed using Ingenuity® Pathway Analysis (IPA). UMD Bos taurus 3.1.1 was used as the comparison genome for both cases since the buffalo genome is not yet fully sequenced. It was obtained from a list of differentially expressed genes of the preovulatory follicles of each species and this difference was quantified, they were grouped according to their biological function and potential upstream regulators. The information was processed individually and compared between the two species. The program works by assigning a probabilistic value to the association between over- or under-expressed genes and major biological functions. For the analysis of upstream regulators, IPA® compares the results of the experiment with its own database of the known effects of genes and molecules on expression. Two values are calculated from each regulator, an overlap value (overlap p) and an activation value (Z score), which corresponds to a calculated numerical average of the known effects of the molecule or gene (up or down regulation) and their respective targets. A Z value is considered significant when it has a value greater than 2 (Z-score > 2) for activation and (Z-score < 2) for inhibition, intermediate values are considered not to be attributable to the experimentation14.

Twenty-one reports on gene expression in bovine granulosa cells in which there was at least one database deposited were found, and of these, only one had information on buffaloes. There were no reports on the comparison of differential gene expression between buffaloes and cows. Finally, the requirements were met by two reports: “Transcriptome profiling of granulosa cells of bovine ovarian follicles during growth from small to large antral sizes”, access number GSE39589 with four repetitions19, and “Buffalo Gene expression profiling of preovulatory follicle in the buffalo cow: effects of increased IGF-I concentration on periovulatory events”, access number GSE11312, with three repetitions18.

In the initial works, it is reported that, in buffaloes, there are 110 differentially expressed genes that belong to 14 metabolic pathways according to the Gene Ontology database, for cows, 446 genes belonging to 10 metabolic pathways were found.

The IPA analysis shows that the most important canonical metabolic pathways associated with the expression pattern observed in the buffalo were: Protein ubiquitination, mitochondrial dysfunction, oxidative phosphorylation and signaling associated with the estrogen receptor and sirtuins, in cows, the most important canonical pathways are the synthesis of triacylglycerides, signal translator and activator of transcription 3 (STAT3), phagosome maturation, Janus Kinase 2 (JAK 2) Protein in the signaling of cytokine-like hormones and that of oncostatin (Table 1).

Table 1 Main metabolic pathways in the buffalo and cow follicle 

Pathway p-value Overlap (%) Molecules
Buffalo follicle
Protein ubiquitination 1.11 E -11 19.70 53/269
Mitochondrial dysfunction 2.41 E-11 22.30 42/188
Oxidative phosphorylation 7.80 E-10 25.20 30/119
Signals of the estrogen receptor 9.46 E -10 23.90 32/134
Sirtuin signaling 3.46 E -08 16.00 52/325
Cow follicle
Triacylglycerol biosynthesis 1.41 E-03 6.90 4/58
STAT3 pathway 1.83 E -03 4.80 5/104
Phagosome maturation 1.97 E-03 3.90 6/155
Role of JAK2 signaling* 2.84 E -03 8.80 .3/34
Oncostatin M signaling 4.53 E-03 7.50 .3/40

*cytokine-like hormone.

The IPA analysis showed that the genes or molecules of the following pathways: HNF4A, RICTOR, EIF4E, 1-2-dithiole-3-thione, STI926 were activated in buffaloes, without being any characterized in buffaloes, and for cows, they were: transforming factor beta 1 (TGFB1), Estrogen receptor type B2 (ERBB2), dexamethasone, beta-estradiol, D-glucose (Table 2).

Table 2 Main upstream regulators 

p-value
Buffaloes
HNF4A 2.70 E-27
RICTOR 2.15 E -17
EIF4E 1.38 E -15
1,2-dithiole-3-thione 4.9 E-15
ST1926 5.64 E-15
Cows
TGFB1 7.64 E-09
ERBB2 1.2 E -08
Dexamethasone 1.3 E E-07
beta-estradiol 5.21 E-07
D-glucose 2.10 E -06

The most important functional networks for buffaloes were: 1) molecular transport, RNA trafficking and post-translational modifications, 2) cell signaling, post-translational modifications and protein synthesis, 3) cell assembly and organization, developmental and hereditary disorders, 4) post-transcriptional RNA modifications, protein synthesis and gene expression, cell maintenance and function, post-translational modifications, and protein folding. For cows 1) cell organization and assembly, developmental disorders and neurological diseases, 2) lipid metabolism, small molecule biochemistry, intracellular signaling and interaction, 3) cell death and survival, interaction between them and cancer, 4) molecular transport, cell signaling and vitamin and mineral metabolism (Table 3).

Table 3 Most important functional networks 

Name of networks and their functions Score
Buffaloes
Molecular transport, RNA trafficking and post-translational modifications 153
Cell signaling, post-translational modifications and protein synthesis 146
Cell assembly and organization, developmental and hereditary disorders 141
Post-transcriptional RNA modifications, protein synthesis and gene expression 139
Cell maintenance and function, post-translational modifications and protein folding 123
Cows
Name of networks and their functions Score
Cell organization and assembly, developmental disorders and neurological diseases 163
Lipid metabolism, small molecule biochemistry, intracellular signaling and interaction 79
Cell death and survival, interaction between them and cancer 73
Molecular transport, cell signaling and vitamin and mineral metabolism 11

In this work, gene expression in preovulatory follicles between two closely related species was compared, the way to do it is a novel approach to have more knowledge. At first glance, there is no strict relationship between the concepts enunciated in the introduction and the results of the experiment. This fact can be considered to be discussed because before doing the analysis, the thought of the researchers was focused on the existing knowledge about endocrine control and follicle development in the species and the results show information associated with aspects related almost exclusively to cell and molecular biology.

It is important to highlight the difficulty in finding comparable information, and given the biological approach of the writing, it is decided to avoid the technical discussion on the way of obtaining the data and its analysis, which is so frequent in this type of publications. In a previous work of the group20, where the information reported in the two articles evaluated in this study is described, it is shown that in the preovulatory follicles of cows and buffaloes, there is only coincidence in the expression of three genes (20 %): tissue plasminogen activator (PLAT), steroidogenic acute regulator, (STAR), coagulation factor II receptor-like I (F2RL-1), with differences in expression levels (relative expression) 9.7 vs 17.5, 8.74 vs 3.4, and 7.7 vs 5.3 times for PLAT, STAR and F2RL-1 respectively, showing the differences between species. Additionally, it has not been reported that any of the genes found have to do directly with ovulation, follicular development or that they are markers of the same biological processes.

PLAT is a secreted serine protease that converts the proenzyme plasminogen into plasmin, a fibrinolytic protein. An increase in its function causes hyperfibrinolysis, which is evidenced by excessive bleeding, and a decrease in its function causes hypofibrinolysis, which is evidenced by thrombosis and embolism, it has been reported overexpressed in granulosa cells of ovulatory follicles and it has been associated with follicular rupture21.

STAR has an important role in the acute regulation of steroid hormone synthesis, allows the breakdown of cholesterol to pregnenolone to facilitate its transport from the outer membrane to the inner one in the mitochondria. In buffaloes, it has been reported that the expression is increased (up regulated) in granulosa cells and follicular wall after the bovine growth hormone treatment18, additionally, a synergism between insulin-like growth factor 1 and gonadotropins to increase the STAR expression22.

F2RL-1 is a protein that belongs to the family of those that are associated with the type I receptor of G proteins, also has a function on the inflammatory response, in innate and adaptive immunity23. Signaling through this gene (F2RL-1) mediates the in vitro cytogenesis of endothelial cells and promotes vasodilation and microvascular permeability, fundamental steps of angiogenesis. Overexpression of the F2RL-1 gene has been observed, partly explaining the growth changes of the blood vessels of the theca cells, and the invasion of the mural granulosa cells after follicular rupture24.

Although there is a low correspondence between the two species, common genes are associated with ovulation mechanisms, genes associated with the way the cell performs its function. No differentially expressed genes are observed associated with the phenomena that direct the process, such as gonadotropins or sex steroids. Consequently, a new way of viewing and analyzing the data obtained must be found since what was found is rather the description of how a phenomenon is executed, which, in this case, would be the proteolytic factors for the rupture of the follicle, the changes in the vasculature for the remodeling of the organ and the accumulation of estrogens to generate the LH peak. For some authors, ovulation has been associated with inflammation25, it could then be assumed that the two species do the same, with a group of general master molecules and another of effectors that can be very specific and mark the differences observed.

Protein ubiquitination is the most important pathway for the degradation of half-life regulatory proteins. Hatzirodos et al20 found that this pathway is involved in the transition from a small to large follicle. Other researchers found it as a regulator of androgen receptors in humans26.

Mitochondria are the main oxygen consumers of the cell; the effect of the proper functioning of the pathway on oocyte competition for its role in the formation of reactive oxygen species has been reported27. In women, mitochondrial transfer has been proposed as an alternative for the treatment of oocytes from patients with diseases that include alterations in the mitochondria28, also in obese humans, that mitochondrial dysfunction is associated with alterations in fertility. In buffaloes, it has been reported that supplementation of maturation media to produce embryos in vitro, with cystine with or without cysteamine significantly increases the proportion of oocytes exhibiting normal fertilization, cleavage and blastocyst production29.

Oxidative phosphorylation is the process by which the cell produces ATP. Li et al6 reported, in buffaloes, that oxidative phosphorylation is important for the early stages of follicular development. Its role in the ovulatory process given the hypoxic nature of the process has also been reported. Evidence that the follicle needs to produce more molecules for the hypoxic response in the preovulatory antral follicles has been shown 5.

Sirtuins belong to class III of deacetylase enzymes that use NAD+ as a substrate to perform their function, it has been reported that there are 7 in mammals with multiple functions (SIRT 1-7), they have function as regulators of transcription in aging, metabolism, cancer, inflammation, DNA repair and cellular response to stress30.

SIRT1, SIRT2 and SIRT3 have been reported to have protective function for oocyte aging after ovulation, in addition, SIRT3 has a role in the detection of reactive oxygen species (ROS) in folliculogenesis and luteinization processes of granulosa cells31. Pacella-Ince et al have described that low mRNA levels of mitochondrial SIRT 5 are associated with reduced ovarian reserve and ovarian aging in humans32.

Most important metabolic pathways in cows:

Triacylglycerol biosynthesis. The effect of lipid droplets on oocyte quality and function is widely documented in the literature33. Today, they are considered as a separate organelle with their own associated enzymes. In humans, the lipid profile of granulosa cells has been successfully associated in gestation, in cow oocytes, it has been associated with quality, and in humans with effect on maturation34.

Phagosome maturation is a process in which some particles that are internalized move on acidified structures, which are integrated into a mechanism of apoptosis. This process necessarily involves maturation, which can be seen by the sequential fusion of the different stages of lysosomes that end the formation of a phagolysosome. There is very little information about the role of this pathway in follicle formation or oocyte quality; it has been suggested as an alternative pathway for the control of oocyte death and that it can be activated during follicle formation for autophagy processes. Alteration of some genes encoding proteins for autophagy cause a large loss of oocytes, suggesting a role of autophagy in regulating their survival35.

Janus kinases are a family of 4 tyrosine kinases. These play an important role in cell growth, survival and differentiation; they are widely expressed in the preovulatory follicle and their expression is inhibited by intrafollicular injection, it has also been reported that in granulosa cells, IL-6 promotes FSH-induced VEGF expression by the JAK/STAT3 pathway36.

Cytokines are the main intercellular mediators, these activate in the ovary immune and non-immune cells that facilitate the constant reorganization of the ovarian stroma through proteolytic pathways, facilitating modifications of the basement membrane of granulosa cells, and endothelial vascular invasion. Martins et al reported that receptors for oncostatin M are regulated in granulosa cells during follicular atresia, ovulation, and luteolysis, and that oncostatin protein from other cells regulate the function of cell viability genes in granulosa and luteal cells37. Many molecules involved in the follicular development that affect signals for ovulation trigger the expression of cytokines such as TNFa, interleukin 7, and CDKN1A38.

When the comparison of gene expression between the two species is made and the 10 activated canonical pathways with the highest P value are analyzed, only a few are shared with the individual analyses, in the case of buffalo, 5 of the 10, while for cows only 1 (Table 4).

Table 4 Comparison of the main metabolic pathways in the two species 

Metabolic pathways p-value Relationship Score- z
Sirtuins signaling pathway 5.02E+00 1.63E-01 2.191
Role of CHK proteins. Proteins in cell cycle control 4.74E+00 2.78E-01 1.897
Oxidative phosphorylation 4.61E+00 2.19E-01 -4.583
De novo biosynthesis of pyridine ribonucleotides 4.38E+00 2.89E-01 -1.941
Mitotic role of the Polo-like kinases 4.07E+00 2.46E-01 -1.667
Interconversion of pyrimidine ribonucleotides 3.93E+00 2.79E-01 -1.732
Androgen signaling 3.85E+00 1.83E-01 0.378
3-phosphoinositide degradation 3.76E+00 1.74E-01 -1.569
Regulation of eIF4 and p70S6K signals 3.67E+00 1.72E-01 0.632
Biosynthesis of D-myo-inositol (1,4,5,6)-tetrakisphosphate 3.65E+00 1.78E-01 -1.225

Upstream regulators in buffaloes:

Hepatocyte nuclear factor 4A (HNF4A) is involved in gluconeogenesis and lipid metabolism. This factor has been reported by Khan et al11 as an upstream regulator in cow granulosa cells after being stimulated with FSH. RIPTOR is a gene that produces a molecule that accompanies the target of rapamycin. In human cells, it has been reported that an mTOR-associated kinase is necessary for Ser473 to be phosphorylated, inhibiting the AkT/PKB metabolic pathway that is involved in apoptosis and oocyte maturation.

The translation initiation factor, EIF4E, produces a protein that helps the start of translation by recruiting ribosomes at the 5’-end of the subunit, limiting the start of translation. Its function is reported to be increased in the transition from primordial follicle to primary follicle39, in the oocyte for the initiation of mRNA translation associated with the continuation of meiosis, and in the inhibition of the TORC1 function for the consumption of amino acids.

IPA also reports on the role of some regulatory molecules of gene function, evaluates genes associated with function control such as 1-2-dithiole-3-thione, which has effect on antioxidant enzymes, and ST1926, which is a retinoic acid-like molecule with effects on growth and differentiation40. There are no reports of the action of these molecules on the function of granulosa cells.

Upstream regulators in cows:

The transforming factor beta-1 belongs to the superfamily of proteins of the transforming factors beta, they have been described in many regulatory activities in the ovary, follicular development and ovulation in different species. Landry et al report that it has a role in the production of a competent oocyte and in the induction of atresia in the case of persistent follicles10.

ERBB2 encodes a tyrosine kinase that belongs to the family of epidermal growth receptors, amplification or overexpression of this gene has been associated with breast and ovarian tumors30 and follicular development19, in the regulation of the expression of the TP53 tumor suppression gene during the proliferation of granulosa cells under the stimulation of FSH and after the LH peak41.

Estrogens, Dexamethasone and D-Glucose are molecules reported with effects on follicular development in cattle42. Estrone and estradiol are steroid hormones synthesized in the ovary, critical for reproductive function, the main enzyme of this pathway is the aromatase CPY19A143.

At this point, it is easily understandable that, despite being bovines, each species has its different way of performing the phenomenon, and that is one of the important results of this work, the evidence shows that each species develops the follicle in a different way. Since it is observed that each species ovulates with different events, the objective of explaining the reproductive differences in reproductive behavior between these two species cannot be met.

The high correspondence of the result obtained when analyzing the most frequent biological networks or functions in the studied event between the two species analyzed, it is shown that both are having the same biological event: growth of a structure within the organism with high metabolic production, cell proliferation, production of signals for all the events that will happen, whose consequence is ovulation. The comparison between networks shows how they do the same with a different number of molecules, buffaloes involve more molecules than cows, which could give them an advantage for ovulation, having more options of cellular pathways available. In the comparative analysis between species, buffaloes do not only share the pathway of lipid metabolism with cows, suggesting that there should be differences at this level in follicular fluid or oocytes, with no reports to date in the literature.

Only a few publications in reproduction have tried to make comparisons between buffaloes and cows with a global approach to the problem, it is interesting to see how the evidence of the differences is increasing. Reports on differences in specific genes in specific events, such as signaling for meiosis restart44, or the role of transforming factors on follicular development45, can easily be found. Much information is known about the physiology of granulosa cells and their role in follicular development in cows, but it is scarce in buffaloes and much less its comparison. There is only one article in the study of the transcriptomics of buffalo granulosa cells, using RNA-seq and slaughter plant material, finding differential expression in 595 genes when comparing the initial states with the end states of follicular development5.

The comparison between species is a novel approach in the area of reproductive biology, studying the same phenomenon on equal conditions between buffaloes and cows shows how each species has developed its own way of carrying out its processes, that the regulation of the same have different pathways between Bos indicus and Bubalus bubalis, so the phenomena must be studied in a particular way for each species and that the extrapolation of the information obtained between species should be avoided and be the basis for the analysis of the complexity of the phenomena studied, with the obligation to see them in a different, non-reductionist way.

Acknowledgements

The authors would like to express their gratitude to the National University of Colombia-Medellín campus and the Colombian Association of Buffalo Breeders for the financing of this work.

REFERENCES

1. Buntjer JB, Otsen M, Nijman IJ, Kuiper MT, Lenstra JA. Phylogeny of bovine species based on AFLP fingerprinting. Heredity 2002;88(1):46-51. [ Links ]

2. Berdugo J, Posada I, Angel J. Need of reevaluation of the parameters of semen straws to be used in artificial insemination programs. Ital J Anim Sci 2007;6(suppl 2):619-21. [ Links ]

3. Drost M. Bubaline versus bovine reproduction. Theriogenology 2007;68(3):447-449. [ Links ]

4. Santos SS, Ferreira MA, Lima MY, Sampaio RV, Cordeiro MS, Silva TV, et al. Quantification, morphology and ultrastructure of preantral follicles of buffalo (Bubalus bubalis) foetuses. Reprod Dom Anim 2011;46(1):e17-22. [ Links ]

5. Gimenes LU, Fantinato Neto P, Arango JS, Ayres H, Baruselli PS. Follicular dynamics of Bos indicus, Bos taurus and Bubalus bubalis heifers treated with norgestomet ear implant associated or not to injectable progesterone. Anim Reprod 2009;6(1):256. [ Links ]

6. Li J, Liu J, Campanile G, Plastow G, Zhang C, Wang Z, et al. Novel insights into the genetic basis of buffalo reproductive performance. BMC Genom 2018;19(1):1-1. [ Links ]

7. Blondin, P, Vigneault, C, Nivet, AL, Sirard, MA. Improving oocyte quality in cows and heifers-What have we learned so far? Anim Reprod 2018;9(3):281-290. [ Links ]

8. Baldrighi JM, Sá Filho MF, Batista EOS, Lopes RN, Visintin JA, Baruselli PS, et al. Anti-mullerian hormone concentration and antral ovarian follicle population in Murrah heifers compared to Holstein and Gyr kept under the same management. Reprod Domest Anim 2014;49(6):1015-1020. [ Links ]

9. Niet AL, Bunel A, Labrecque R, Belanger J, Vigneault C, Blondin P, et al. FSH withdrawal improves developmental competence of oocytes in the bovine model. Reproduction 2012;143(2):165-171. [ Links ]

10. Carvalho NA, Soares JG, Porto Filho RM, Gimenes LU, Souza DC, Nichi M, et al. Equine chorionic gonadotropin improves the efficacy of a timed artificial insemination protocol in buffalo during the nonbreeding season. Theriogenology 2013;79(3):423-428. [ Links ]

11. Khan DR, Landry DA, Fournier É, Vigneault C, Blondin P, Sirard MA. Transcriptome meta-analysis of three follicular compartments and its correlation with ovarian follicle maturity and oocyte developmental competence in cows. Physiol Genom 2016;48(8):633-643. [ Links ]

12. Fu Q, Huang Y, Wang Z, Chen F, Huang D, Lu Y, et al. Proteome profile and quantitative proteomic analysis of buffalo (Bubalus bubalis) follicular fluid during follicle development. Int J Mol Sci 2016;17(5):618. [ Links ]

13. Mondadori RG, Luque MC, Santin TR, Báo SN. Ultrastructural and morphometric characterization of buffalo (Bubalus bubalis) ovarian preantral follicles. Anim Rep Sci 2007;97(3-4):323-333. [ Links ]

14. Robert C. Microarray analysis of gene expression during early development: A cautionary overview. Reproduction 2010;140(6);787-801. [ Links ]

15. Krämer A, Green J, Pollard Jr J, Tugendreich S. Causal analysis approaches in ingenuity pathway analysis. Bioinformatics 2014;30(4):523-530. [ Links ]

16. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science 2006;313(5795):1929-1935. [ Links ]

17. Abatangelo L, Maglietta R, Distaso A, D'Addabbo A, Creanza TM, Mukherjee S, et al. Comparative study of gene set enrichment methods. BMC Bioinfo 2009;10(1):1-2. [ Links ]

18. Pollard Jr J, Butte AJ, Hoberman S, Joshi M, Levy J, Pappo J. A computational model to define the molecular causes of type 2 diabetes mellitus. Diab Tech Ther 2005;7(2):323-336. [ Links ]

19. Rao JU, Shah KB, Puttaiah J, Rudraiah M. Gene expression profiling of preovulatory follicle in the buffalo cow: effects of increased IGF-I concentration on periovulatory events. PloS One 2011;6(6):e20754. [ Links ]

20. Hatzirodos N, Hummitzsch K, Irving-Rodgers HF, Harland ML, Morris SE, Rodgers RJ. Transcriptome profiling of granulosa cells from bovine ovarian follicles during atresia. BMC Genom 2014;15(1):1-26. [ Links ]

21. Berdugo JA, Echeverri JJ, Tarazona AM, López A. Differences in transcriptomic data from preovulatory follicles of buffalo (Bubalus bubalis) and (Bos indicus) cattle: A meta-analysis No Title. Anim Reprod 2018;15(Suppl 1):1161. [ Links ]

22. Lussier JG, Diouf MN, Lévesque V, Sirois J, Ndiaye K. Gene expression profiling of upregulated mRNAs in granulosa cells of bovine ovulatory follicles following stimulation with hCG. Reprod Biol Endocrinol 2017;15(1):1-6. [ Links ]

23. Natesampillai S, Kerkvliet J, Leung PC, Veldhuis JD. Regulation of Kruppel-like factor 4, 9, and 13 genes and the steroidogenic genes LDLR, StAR, and CYP11A in ovarian granulosa cells. Am J Physiol-End Metab 2008;294(2):E385-91. [ Links ]

24. Castilho AC, Dalanezi FM, Franchi FF, Price CA, Ferreira JC, Trevisol E, et al. Expression of fibroblast growth factor 22 (FGF22) and Its receptor, FGFR1B, during development and regression of bovine corpus luteum. Theriogenology 2019;125:1-5. [ Links ]

25. Harlow CR, Hillier SG. Connective tissue growth factor in the ovarian paracrine system. Mol Cell End 2002;187(1-2):23-27. [ Links ]

26. Ruebel ML, Cotter M, Sims CR, Moutos DM, Badger TM, Cleves MA, et al. Obesity modulates inflammation and lipid metabolism oocyte gene expression: a single-cell transcriptome perspective. J Clin End Metab 2017;102(6):2029-38. [ Links ]

27. Lim JJ, Lima PD, Salehi R, Lee DR, Tsang BK. Regulation of androgen receptor signaling by ubiquitination during folliculogenesis and its possible dysregulation in polycystic ovarian syndrome. Scientific Reports 2017;31;7(1):1-2. [ Links ]

28. Benkhalifa M, Ferreira YJ, Chahine H, Louanjli N, Miron P, Merviel P, et al. Mitochondria: participation to infertility as source of energy and cause of senescence. Int J Biochem Cell Biol 2014;55:60-64. [ Links ]

29. Cecchino GN, Seli E, Da Motta EL, García-Velasco JA. The role of mitochondrial activity in female fertility and assisted reproductive technologies: overview and current insights. Reprod Biomed Online 2018;36(6):686-697. [ Links ]

30. Gasparrini B, Boccia L, Marchandise J, Di Palo R, George F, Donnay I, et al. Enrichment of in vitro maturation medium for buffalo (Bubalus bubalis) oocytes with thiol compounds: Effects of cystine on glutathione synthesis and embryo development. Theriogenology 2006;65(2):275-287. [ Links ]

31. Li J, Li Z, Liu S, Zia R, Liang A, Yang L. Transcriptome studies of granulosa cells at different stages of ovarian follicular development in buffalo. Anim Reprod Sci 2017;(187):181-192. [ Links ]

32. Sirotkin AV. The role and application of sirtuins and mTOR signaling in the control of ovarian functions. Cells 2016;5(4):42. [ Links ]

33. Fu H, Wada-Hiraike O, Hirano M, Kawamura Y, Sakurabashi A, Shirane A, et al. SIRT3 positively regulates the expression of folliculogenesis-and luteinization-related genes and progesterone secretion by manipulating oxidative stress in human luteinized granulosa cells. Endocrininol 2014;155(8):3079-3087. [ Links ]

34. Pacella-Ince L, Zander-Fox DL, Lane M. Mitochondrial SIRT5 is present in follicular cells and is altered by reduced ovarian reserve and advanced maternal age. Reprod Fert Develop 2014;26(8):1072-1083. [ Links ]

35. Dunning KR, Russell DL, Robker RL. Lipids and oocyte developmental competence: the role of fatty acids and b-oxidation. Reproduction 2014;148(1):R15-27. [ Links ]

36. Fayezi S, Ghaffari-Novin M, Darabi M, Norouzian M, Nouri M, Farzadi L, et al. Primary culture of human cumulus cells requires stearoyl-coenzyme a desaturase 1 activity for steroidogenesis and enhancing oocyte in vitro maturation. Reprod Sci 2018;(6):844-853. [ Links ]

37. Gawriluk TR, Hale AN, Flaws JA, Dillon CP, Green DR, Rucker EB. Autophagy is a cell survival program for female germ cells in the murine ovary. Reproduction 2011;141(6):759. [ Links ]

38. Yang M, Wang L, Wang X, Wang X, Yang Z LJ. IL-6 promotes FSH-induced VEGF expression through JAK/STAT3 signaling pathway in bovine granulosa cells. Cell Physiol Biochem 2017;44(1):293-302. [ Links ]

39. Martins KR, Haas CS, Ferst, JG, Rovani, MT, Goetten AL, Duggavathi R. et al. Oncostatin M and its receptors mRNA regulation in bovine granulosa and luteal cells. Theriogenology 2019;125;324-330. [ Links ]

40. Stassi AF, Baravalle ME, Belotti EM, Rey F, Gareis NC, Díaz PU, et al. Altered expression of cytokines IL-1α, IL-6, IL-8 and TNF-α in bovine follicular persistence. Theriogenology 2017;97:104-112. [ Links ]

41. Ernst EH, Grøndahl ML, Grund S, Hardy K, Heuck A, Sunde L, Franks S, et al. Dormancy and activation of human oocytes from primordial and primary follicles: molecular clues to oocyte regulation. Hum Reprod 2017;32(8):1684-1700. [ Links ]

42. El Hajj H, Khalil B, Ghandour B, Nasr R, Shahine S, Ghantous A, et al. Preclinical efficacy of the synthetic retinoid ST1926 for treating adult T-cell leukemia/lymphoma. Blood 2014;124(13):2072-2080. [ Links ]

43. Sirotkin AV, Ovcharenko D, Benčo A, Mlynček M. Protein kinases controlling PCNA and p53 expression in human ovarian cells. Funct Integr Genom 2009;(2):185-195. [ Links ]

44. Noma N, Kawashima I, Fan HY, Fujita Y, Kawai T, Tomoda Y, et al. LH-induced neuregulin 1 (NRG1) type III transcripts control granulosa cell differentiation and oocyte maturation. Mol End 2011;25(1):104-116. [ Links ]

45. Baufeld A, Vanselow J. A tissue culture model of estrogen-producing primary bovine granulosa cells. Journal of visualized experiments: JoVE 2018;(139):58208. [ Links ]

Received: February 25, 2020; Accepted: December 22, 2020

Creative Commons License Este es un artículo publicado en acceso abierto bajo una licencia Creative Commons