Animal Reproduction (AR)
https://www.animal-reproduction.org/article/doi/10.1590/1984-3143-AR2020-0057
Animal Reproduction (AR)
Thematic Section: 34th Annual Meeting of the Brazilian Embryo Technology Society (SBTE)

New approaches to diagnose and target reproductive failure in cattle

Ky Garrett Pohler; Sydney Taylor Reese; Gessica Araujo Franco; Ramiro Vander Oliveira Filho; Rafael Paiva; Lohana Fernandez; Gabriela de Melo; José Luiz Moraes Vasconcelos; Reinaldo Cooke; Rebecca Kyle Poole

Downloads: 2
Views: 704

Abstract

Abstract: Reproductive failure and pregnancy loss in cattle are some of the largest economic burdens to cattle producers and one of most perplexing factors influencing management decisions. Pregnancy loss may occur at any point during gestation with the largest percentage of loss occurring in the first 30 days and, subsequently, decreasing as the pregnancy progresses. Losses may be attributed to numerous factors, predisposed issues or environmental conditions such as nutritional stressors or disease. From a research perspective, determining the exact causes of pregnancy loss or embryonic mortality in cattle have been difficult, due to limitations of accurately determining early gestation pregnancy status. Until methods that precisely determine embryo success early in gestation are available, our understanding of in vivo pregnancy loss will lack clarity necessary to develop management strategies to decrease such loss. In this review, we will briefly discuss the pivotal periods of pregnancy loss affecting beef and dairy cattle, methods and technologies to determine pregnancy status and embryo viability and potential opportunities to decrease reproductive failure.

Keywords

pregnancy, bovine, embryonic mortality, pregnancy loss, pregnancy detection

References

Andersen K, LeFever D, Brinks J, Odde K. The use of reproductive tract scoring in beef heifers. Agri-Practice. 1991;12:106-11.

Arnold H, Martins JP, Oliveira LZ, Policelli R, Stomack K, Sasser G, Pursley J. Effectiveness of pregnancy-specific protein B in pregnancy diagnosis of dairy cows and heifers. J Dairy Sci. 2012;95(Suppl. 1):128.

Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Gibson DF, Mitchell PS, Bennett CF, Pogosova-Agadjanyan EL, Stirewalt DL, Tait JF, Tewari M. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci USA. 2011;108(12):5003-8. http://dx.doi.org/10.1073/pnas.1019055108. PMid:21383194.

Assis AC No, Pereira F, Santos TC, Ambrosio CE, Leiser R, Miglino MA. Morpho‐physical recording of bovine conceptus (Bos indicus) and placenta from days 20 to 70 of pregnancy. Reprod Domest Anim. 2010;45(5):760-72. PMid:19281595.

Austin KJ, King CP, Vierk JE, Sasser RG, Hansen TR. Pregnancy-specific protein B induces release of an alpha chemokine in bovine endometrium. Endocrinology. 1999;140(1):542-5. http://dx.doi.org/10.1210/endo.140.1.6608. PMid:9886868.

Baez GM, Barletta RV, Guenther JN, Gaska JM, Wiltbank MC. Effect of uterine size on fertility of lactating dairy cows. Theriogenology. 2016;85(8):1357-66. http://dx.doi.org/10.1016/j.theriogenology.2015.04.022. PMid:26924681.

Bai H, Sakurai T, Godkin JD, Imakawa K. Expression and potential role of GATA factors in trophoblast development. J Reprod Dev. 2013;59:1– 6. https://doi.org/10.1262/jrd.2012-100. PMid:23428586

Balaro MFA, Santos AS, Moura LFG, Fonseca JF, Brandão FZ. Luteal dynamic and functionality assessment in dairy goats by luteal blood flow, luteal biometry, and hormonal assay. Theriogenology. 2017;95:118-26. http://dx.doi.org/10.1016/j.theriogenology.2017.02.021. PMid:28460665.

Ball L, Carroll E. Induction of fetal death in cattle by manual rupture of the amniotic vesicle. J Am Vet Med Assoc. 1963;142:373-4. PMid:13966137.

Barton SC, Adams CA, Norris M, Surani M. Development of gynogenetic and parthenogenetic inner cell mass and trophectoderm tissues in reconstituted blastocysts in the mouse. J Embryol Exp Morphol. 1985;90(1):267-85. PMid:3834032.

Bazer FW, Spencer TE, Ott TL. Interferon tau: a novel pregnancy recognition signal. Am J Reprod Immunol. 1997;37(6):412-20. http://dx.doi.org/10.1111/j.1600-0897.1997.tb00253.x. PMid:9228295.

Bazer FW, Ying W, Wang X, Dunlap KA, Zhou B, Johnson GA, Wu G. The many faces of interferon tau. Amino Acids. 2015;47(3):449-60. http://dx.doi.org/10.1007/s00726-014-1905-x. PMid:25557050.

Beal WE, Perry RC, Corah LR. The use of ultrasound in monitoring reproductive physiology of beef cattle. J Anim Sci. 1992;70(3):924-9. http://dx.doi.org/10.2527/1992.703924x. PMid:1564011.

Bem THC, Silveira JC, Sampaio RV, Sangalli JR, Oliveira MLF, Ferreira RM, Silva LA, Perecin F, King WA, Meirelles FV, Ramos ES. Low levels of exosomal-miRNAs in maternal blood are associated with early pregnancy loss in cloned cattle. Sci Rep. 2017;7(1):14319. http://dx.doi.org/10.1038/s41598-017-14616-1. PMid:29085015.

Bó GA, Cedeño A. Expression of estrus as a relevant factor in fixed-time embryo transfer programs using estradiol/progesterone-based protocols in cattle. Anim Reprod. 2018;15(3):224-30. http://dx.doi.org/10.21451/1984-3143-AR2018-0060.

Bott RC, Ashley RL, Henkes LE, Antoniazzi AQ, Bruemmer JE, Niswender GD, Bazer FW, Spencer TE, Smirnova NP, Anthony RV, Hansen TR. Uterine vein infusion of interferon tau (IFNT) extends luteal life span in ewes. Biol Reprod. 2010;82(4):725-35. http://dx.doi.org/10.1095/biolreprod.109.079467. PMid:20042537.

Breukelman S, Perényi Z, Taverne M, Jonker H, Van der Weijden G, Vos P, de Ruigh L, Dieleman S, Beckers J-F, Szenci O. Characterisation of pregnancy losses after embryo transfer by measuring plasma progesterone and bovine pregnancy-associated glycoprotein-1 concentrations. Vet J. 2012;194(1):71-6. http://dx.doi.org/10.1016/j.tvjl.2012.02.020. PMid:22516919.

Burns G, Brooks K, Wildung M, Navakanitworakul R, Christenson LK, Spencer TE. Extracellular vesicles in luminal fluid of the ovine uterus. PLoS One. 2014;9(3):e90913. http://dx.doi.org/10.1371/journal.pone.0090913. PMid:24614226.

Butler J, Hamilton W, Sasser R, Ruder C, Hass G, Williams R. Detection and partial characterization of two bovine pregnancy-specific proteins. Biol Reprod. 1982;26(5):925-33. http://dx.doi.org/10.1095/biolreprod26.5.925. PMid:6807365.

Cameron A, Silveira J, Bouma G, Bruemmer J. Evaluation of exosomes containing miRNA as an indicator of pregnancy status in the mare. J Equine Vet Sci. 2011;31(5):314-5. http://dx.doi.org/10.1016/j.jevs.2011.03.148.

Carter F, Forde N, Duffy P, Wade M, Fair T, Crowe M, Evans A, Kenny D, Roche J, Lonergan P. Effect of increasing progesterone concentration from Day 3 of pregnancy on subsequent embryo survival and development in beef heifers. Reprod Fertil Dev. 2008;20(3):368-75. http://dx.doi.org/10.1071/RD07204. PMid:18402756.

Cooke RF. Early career achievement award: supplementing omega-6 fatty acids to enhance early embryonic development and pregnancy establishment in Bos indicus and B. taurus beef cows. J Anim Sci. 2019;97(1):485-95. http://dx.doi.org/10.1093/jas/sky414. PMid:30351357.

Curran S, Pierson R, Ginther O. Ultrasonographic appearance of the bovine conceptus from days 20 through 60. J Am Vet Med Assoc. 1986;189(10):1295-302. PMid:3793570.

Diskin M, Sreenan J. Fertilization and embryonic mortality rates in beef heifers after artificial insemination. J Reprod Fertil. 1980;59(2):463-8. http://dx.doi.org/10.1530/jrf.0.0590463. PMid:7431304.

Etheridge A, Lee I, Hood L, Galas D, Wang K. Extracellular microRNA: a new source of biomarkers. Mutat Res. 2011;717(1-2):85-90. http://dx.doi.org/10.1016/j.mrfmmm.2011.03.004. PMid:21402084.

Fiandanese N, Viglino A, Strozzi F, Stella A, Williams JL, Lonergan P, Forde N, Iamartino D. 71 Circulating microRNAs as potential biomarkers of early pregnancy in high producing dairy cows. Reprod Fertil Dev. 2016;28(2):165. http://dx.doi.org/10.1071/RDv28n2Ab71.

Franco G, Reese S, Poole R, Rhinehart J, Thompson K, Cooke R, Pohler K. Sire contribution to pregnancy loss in different periods of embryonic and fetal development of beef cows. Theriogenology. 2020;154:84-91. http://dx.doi.org/10.1016/j.theriogenology.2020.05.021. PMid:32535394.

Franco GA, Peres RFG, Martins CFG, Reese ST, Vasconcelos JLM, Pohler KG. Sire contribution to pregnancy loss and pregnancy-associated glycoprotein production in Nelore cows. J Anim Sci. 2018;96(2):632-40. http://dx.doi.org/10.1093/jas/sky015. PMid:29518245.

Garrett J, Geisert R, Zavy M, Morgan G. Evidence for maternal regulation of early conceptus growth and development in beef cattle. J Reprod Fertil. 1988;84(2):437-46. http://dx.doi.org/10.1530/jrf.0.0840437. PMid:3199361.

Gatea AO, Smith MF, Pohler KG, Egen T, Pereira MH, Vasconselos JL, Lawrence JC, Green JA. The ability to predict pregnancy loss in cattle with ELISAs that detect pregnancy associated glycoproteins is antibody dependent. Theriogenology. 2018;108:269-76. http://dx.doi.org/10.1016/j.theriogenology.2017.12.021. PMid:29275034.

Geary TW, Burns GW, Moraes JG, Moss JI, Denicol AC, Dobbs KB, Ortega MS, Hansen PJ, Wehrman ME, Neibergs H, O’Neil E, Behura S, Spencer TE. Identification of beef heifers with superior uterine capacity for pregnancy 1. Biol Reprod. 2016;95(2):47. http://dx.doi.org/10.1095/biolreprod.116.141390. PMid:27417907.

Gebremedhn S, Salilew-Wondim D, Hoelker M, Held-Hoelker E, Neuhoff C, Tholen E, Schellander K, Tesfaye D. Exploring maternal serum microRNAs during early pregnancy in cattle. Theriogenology. 2018;121:196-203. http://dx.doi.org/10.1016/j.theriogenology.2018.08.020. PMid:30172131.

Gifford C, Racicot K, Clark D, Austin K, Hansen T, Lucy M, Davies C, Ott T. Regulation of interferon-stimulated genes in peripheral blood leukocytes in pregnant and bred, nonpregnant dairy cows. J Dairy Sci. 2007;90(1):274-80. http://dx.doi.org/10.3168/jds.S0022-0302(07)72628-0. PMid:17183095.

Ginther O, Shrestha H, Fuenzalida M, Shahiduzzaman A, Beg M. Characteristics of pulses of 13, 14-dihydro-15-keto-prostaglandin F2alpha before, during, and after spontaneous luteolysis and temporal intrapulse relationships with progesterone concentrations in cattle. Biol Reprod. 2010;82(6):1049-56. http://dx.doi.org/10.1095/biolreprod.109.081976. PMid:20147732.

Gray C, Burghardt R, Johnson G, Bazer F, Spencer T. Evidence that absence of endometrial gland secretions in uterine gland knockout ewes compromises conceptus survival and elongation. Reproduction. 2002;124(2):289-300. http://dx.doi.org/10.1530/rep.0.1240289. PMid:12141942.

Green J, Okamura C, Poock S, Lucy M. Measurement of interferon-tau (IFN-τ) stimulated gene expression in blood leukocytes for pregnancy diagnosis within 18-20d after insemination in dairy cattle. Anim Reprod Sci. 2010;121(1-2):24-33. http://dx.doi.org/10.1016/j.anireprosci.2010.05.010. PMid:20554404.

Griffiths-Jones S, Saini HK, van Dongen S, Enright AJ. miRBase: tools for microRNA genomics. Nucleic Acids Res. 2008;36(Suppl. 1):D154-8. http://dx.doi.org/10.1093/nar/gkm952. PMid:17991681.

Han H, Austin KJ, Rempel LA, Hansen TR. Low blood ISG15 mRNA and progesterone levels are predictive of non-pregnant dairy cows. J Endocrinol. 2006;191(2):505-12. http://dx.doi.org/10.1677/joe.1.07015. PMid:17088421.

Hata T, Aoki S, Manabe A, Hata K, Miyazaki K. Three-dimensional ultrasonography in the first trimester of human pregnancy. Hum Reprod. 1997;12(8):1800-4. http://dx.doi.org/10.1093/humrep/12.8.1800. PMid:9308815.

Herzog K, Brockhan-Lüdemann M, Kaske M, Beindorff N, Paul V, Niemann H, Bollwein H. Luteal blood flow is a more appropriate indicator for luteal function during the bovine estrous cycle than luteal size. Theriogenology. 2010;73(5):691-7. http://dx.doi.org/10.1016/j.theriogenology.2009.11.016. PMid:20071016.

Hoeben D, Burvenich C, Massart-Leën A-M, Lenjou M, Nijs G, Van Bockstaele D, Beckers J-F. In vitro effect of ketone bodies, glucocorticosteroids and bovine pregnancy-associated glycoprotein on cultures of bone marrow progenitor cells of cows and calves. Vet Immunol Immunopathol. 1999;68(2-4):229-40. http://dx.doi.org/10.1016/S0165-2427(99)00031-8. PMid:10438322.

Holm DE, Thompson PN, Irons PC. The value of reproductive tract scoring as a predictor of fertility and production outcomes in beef heifers. J Anim Sci. 2009;87(6):1934-40. http://dx.doi.org/10.2527/jas.2008-1579. PMid:19286816.

Ioannidis J, Donadeu FX. Circulating miRNA signatures of early pregnancy in cattle. BMC Genomics. 2016;17(1):184. http://dx.doi.org/10.1186/s12864-016-2529-1. PMid:26939708.

Ioannidis J, Donadeu FX. Changes in circulating microRNA levels can be identified as early as day 8 of pregnancy in cattle. PLoS One. 2017;12(4):e0174892. http://dx.doi.org/10.1371/journal.pone.0174892. PMid:28380001.

Ireland J, Zielak-Steciwko A, Jimenez-Krassel F, Folger J, Bettegowda A, Scheetz D, Walsh S, Mossa F, Knight P, Smith G, Lonergan P, Evans AC. Variation in the ovarian reserve is linked to alterations in intrafollicular estradiol production and ovarian biomarkers of follicular differentiation and oocyte quality in cattle. Biol Reprod. 2009;80(5):954-64. http://dx.doi.org/10.1095/biolreprod.108.073791. PMid:19164170.

Jimenez-Krassel F, Folger J, Ireland J, Smith G, Hou X, Davis JS, Lonergan P, Evans A, Ireland J. Evidence that high variation in ovarian reserves of healthy young adults has a negative impact on the corpus luteum and endometrium during estrous cycles in cattle. Biol Reprod. 2009;80(6):1272-81. http://dx.doi.org/10.1095/biolreprod.108.075093. PMid:19211804.

Kähn W. Sonographic fetometry in the bovine. Theriogenology. 1989;31(5):1105-21. http://dx.doi.org/10.1016/0093-691X(89)90494-9. PMid:16726628.

Kastelic J, Curran S, Pierson R, Ginther O. Ultrasonic evaluation of the bovine conceptus. Theriogenology. 1988;29(1):39-54. http://dx.doi.org/10.1016/0093-691X(88)90030-1.

Kizaki K, Shichijo-Kizaki A, Furusawa T, Takahashi T, Hosoe M, Hashizume K. Differential neutrophil gene expression in early bovine pregnancy. Reprod Biol Endocrinol. 2013;11(1):6. http://dx.doi.org/10.1186/1477-7827-11-6. PMid:23384108.

Kunii H, Koyama K, Ito T, Suzuki T, Balboula AZ, Shirozu T, Bai H, Nagano M, Kawahara M, Takahashi M. Hot topic: pregnancy-induced expression of interferon-stimulated genes in the cervical and vaginal mucosal membranes. J Dairy Sci. 2018;101(9):8396-400. http://dx.doi.org/10.3168/jds.2017-14251. PMid:29935833.

Liang J, Wang S, Wang Z. Role of microRNAs in embryo implantation. Reprod Biol Endocrinol. 2017;15(1):90. http://dx.doi.org/10.1186/s12958-017-0309-7. PMid:29162091.

Løvendahl P, Chagunda M. On the use of physical activity monitoring for estrus detection in dairy cows. J Dairy Sci. 2010;93(1):249-59. http://dx.doi.org/10.3168/jds.2008-1721. PMid:20059923.

Lucy M. Reproductive loss in high-producing dairy cattle: where will it end? J Dairy Sci. 2001;84(6):1277-93. http://dx.doi.org/10.3168/jds.S0022-0302(01)70158-0. PMid:11417685.

Madureira A, Silper B, Burnett T, Polsky L, Cruppe L, Veira D, Vasconcelos J, Cerri R. Factors affecting expression of estrus measured by activity monitors and conception risk of lactating dairy cows. J Dairy Sci. 2015;98(10):7003-14. http://dx.doi.org/10.3168/jds.2015-9672. PMid:26254517.

Madureira AML, Franco GA, Guida TG, Edwards JL, Schrick FN, Vasconcelos JLM, Cerri RLA, Pohler KG. Effect of size and position of the reproductive tract on concentrations of bovine Pregnancy Associated Glycoproteins (PAGs) and the relationship with fertility. Washington: Society for the Study of Reproduction; 2017.

Majumder K, Gelbaya TA, Laing I, Nardo LG. The use of anti-Müllerian hormone and antral follicle count to predict the potential of oocytes and embryos. Eur J Obstet Gynecol Reprod Biol. 2010;150(2):166-70. http://dx.doi.org/10.1016/j.ejogrb.2010.02.029. PMid:20223579.

Mann G, Lamming G. The influence of progesterone during early pregnancy in cattle. Reprod Domest Anim. 1999;34(3‐4):269-74. http://dx.doi.org/10.1111/j.1439-0531.1999.tb01250.x.

Markkandan K, Ahn K, Lee DJ, Kim TI, Dang C, Hong S-E, Yoon H-B, Lim H-J, Hong CP. Profiling and identification of pregnancy-associated circulating microRNAs in dairy cattle. Genes Genomics. 2018;40(10):1111-7. http://dx.doi.org/10.1007/s13258-018-0668-2. PMid:30264330.

Martins J, Wang D, Mu N, Rossi G, Martini A, Martins V, Pursley J. Level of circulating concentrations of progesterone during ovulatory follicle development affects timing of pregnancy loss in lactating dairy cows. J Dairy Sci. 2018;101(11):10505-25. http://dx.doi.org/10.3168/jds.2018-14410. PMid:30197145.

Matsuyama S, Kojima T, Kato S, Kimura K. Relationship between quantity of IFNT estimated by IFN-stimulated gene expression in peripheral blood mononuclear cells and bovine embryonic mortality after AI or ET. Reprod Biol Endocrinol. 2012;10(1):21. http://dx.doi.org/10.1186/1477-7827-10-21. PMid:22439976.

McNeel AK, Soares ÉM, Patterson AL, Vallet JL, Wright EC, Larimore EL, Amundson OL, Miles JR, Chase CC Jr, Lents CA, Wood JR, Cupp AS, Perry GA, Cushman RA. Beef heifers with diminished numbers of antral follicles have decreased uterine protein concentrations. Anim Reprod Sci. 2017;179:1-9. http://dx.doi.org/10.1016/j.anireprosci.2017.01.004. PMid:28215453.

Melo GD, Mello BP, Ferreira CA, Godoy CAS Fo, Rocha CC, Silva AG, Reese ST, Madureira EH, Pohler KG, Pugliesi G. Applied use of interferon-tau stimulated genes expression in polymorphonquclear cells to detect pregnancy compared to other early predictors in beef cattle. Theriogenology. 2020a;152:94-105. http://dx.doi.org/10.1016/j.theriogenology.2020.04.001. PMid:32387553.

Melo GD, Pinto LFM, Rocha CC, Motta IG, Silva LA, Silveira JC, Gonella-Diaza AM, Binelli M, Pugliesi G. Type I interferon receptors and interferon-t stimulated genes in peripheral blood mononuclear cells and polymorphonuclear leucocytes in early pregnancy in beef heifers. Reprod Fertil Dev. 2020b;32(11):953-66. http://dx.doi.org/10.1071/RD19430. PMid:32646539.

Middleton E, Pursley J. Blood samples before and after embryonic attachment accurately determine non-pregnant lactating dairy cows at 24 d post-artificial insemination using a commercially available assay for pregnancy-specific protein B. J Dairy Sci. 2019;102(8):7570-5. http://dx.doi.org/10.3168/jds.2018-15961. PMid:31178191.

Mirando M, Short E Jr, Geisert R, Vallet J, Bazer F. Stimulation of 2′, 5′-oligoadenylate synthetase activity in sheep endometrium during pregnancy, by intrauterine infusion of ovine trophoblast protein-1, and by intramuscular administration of recombinant bovine interferon-αI1. J Reprod Fertil. 1991;93(2):599-607. http://dx.doi.org/10.1530/jrf.0.0930599. PMid:1787480.

Momont H. Rectal palpation: safety issues. In: Proceedings of the Dairy Herd Health Programming Conference; 1990; Minnesota. Minnesota: University of Minnesota; 1990. p. 6-7.

Moraes FLZ, Morotti F, Costa CB, Lunardelli PA, Seneda MM. Relationships between antral follicle count, body condition, and pregnancy rates after timed-AI in Bos indicus cattle. Theriogenology. 2019;136:10-4. http://dx.doi.org/10.1016/j.theriogenology.2019.06.024. PMid:31234052.

Moraes JG, Behura SK, Geary TW, Hansen PJ, Neibergs HL, Spencer TE. Uterine influences on conceptus development in fertility-classified animals. Proc Natl Acad Sci USA. 2018;115(8):E1749-58. http://dx.doi.org/10.1073/pnas.1721191115. PMid:29432175.

Mossa F, Walsh S, Butler S, Berry D, Carter F, Lonergan P, Smith G, Ireland J, Evans A. Low numbers of ovarian follicles≥ 3 mm in diameter are associated with low fertility in dairy cows. J Dairy Sci. 2012;95(5):2355-61. http://dx.doi.org/10.3168/jds.2011-4325. PMid:22541464.

Motta IG, Rocha CC, Bisinotto DZ, Melo GD, Ataide GA Jr, Silva AG, Gonzaga VHG, Santos JA, Freitas BG, Lemes KM, Madureira EH. Increased pregnancy rate in beef heifers resynchronized with estradiol at 14 days after TAI. Theriogenology. 2020;147:62-70. http://dx.doi.org/10.1016/j.theriogenology.2020.02.009. PMid:32097817.

Nebel RL, Whittier W, Cassell B, Britt J. Comparison of on-farm and laboratory milk progesterone assays for identifying errors in detection of estrus and diagnosis of pregnancy. J Dairy Sci. 1987;70(7):1471-6. http://dx.doi.org/10.3168/jds.S0022-0302(87)80171-6. PMid:3624599.

Nebel R. On-farm milk progesterone tests. J Dairy Sci. 1988;71(6):1682-90. http://dx.doi.org/10.3168/jds.S0022-0302(88)79733-7. PMid:3403763.

Niswender GD, Juengel JL, Silva PJ, Rollyson MK, McIntush EW. Mechanisms controlling the function and life span of the corpus luteum. Physiol Rev. 2000;80(1):1-29. http://dx.doi.org/10.1152/physrev.2000.80.1.1. PMid:10617764.

Oliveira R Fo, Franco G, Reese S, Dantas F, Fontes P, Cooke R, Rhinehart J, Thompson K, Pohler K. Using pregnancy associated glycoproteins (PAG) for pregnancy detection at day 24 of gestation in beef cattle. Theriogenology. 2020;141:128-33. http://dx.doi.org/10.1016/j.theriogenology.2019.09.014. PMid:31539641.

Oliveira JF, Henkes LE, Ashley RL, Purcell SH, Smirnova NP, Veeramachaneni DR, Anthony RV, Hansen TR. Expression of interferon (IFN)-stimulated genes in extrauterine tissues during early pregnancy in sheep is the consequence of endocrine IFN-τ release from the uterine vein. Endocrinology. 2008;149(3):1252-9. http://dx.doi.org/10.1210/en.2007-0863. PMid:18063687.

Omontese B, Gomes G, Santos A, Silva L, Merenda V, Bisinotto R. Use of on-farm milk progesterone information to predict fertility outcomes in dairy cows subjected to timed artificial insemination. J Dairy Sci. 2020;103(7):6600-11. http://dx.doi.org/10.3168/jds.2019-17405. PMid:32359997.

Ortega MS, Moraes JG, Patterson DJ, Smith MF, Behura SK, Poock S, Spencer TE. Influences of sire conception rate on pregnancy establishment in dairy cattle. Biol Reprod. 2018;99(6):1244-54. http://dx.doi.org/10.1093/biolre/ioy141. PMid:29931362.

Parr M, Mullen M, Crowe M, Roche J, Lonergan P, Evans A, Diskin M. Relationship between pregnancy per artificial insemination and early luteal concentrations of progesterone and establishment of repeatability estimates for these traits in Holstein-Friesian heifers. J Dairy Sci. 2012;95(5):2390-6. http://dx.doi.org/10.3168/jds.2011-4498. PMid:22541467.

Pereira M, Wiltbank M, Vasconcelos J. Expression of estrus improves fertility and decreases pregnancy losses in lactating dairy cows that receive artificial insemination or embryo transfer. J Dairy Sci. 2016;99(3):2237-47. http://dx.doi.org/10.3168/jds.2015-9903. PMid:26723130.

Pierson R, Ginther O. Ultrasonography for detection of pregnancy and study of embryonic development in heifers. Theriogenology. 1984;22(2):225-33. http://dx.doi.org/10.1016/0093-691X(84)90435-7. PMid:16725953.

Pohler KG, Geary TW, Johnson CL, Atkins JA, Jinks EM, Busch DC, Green JA, MacNeil MD, Smith MF. Circulating bovine pregnancy associated glycoproteins are associated with late embryonic/fetal survival but not ovulatory follicle size in suckled beef cows. J Anim Sci. 2013;91(9):4158-67. http://dx.doi.org/10.2527/jas.2013-6348. PMid:23825331.

Pohler KG, Green JA, Geary TW, Peres RF, Pereira MH, Vasconcelos JL, Smith MF. Predicting embryo presence and viability. Adv Anat Embryol Cell Biol. 2015;216:253-70. http://dx.doi.org/10.1007/978-3-319-15856-3_13. PMid:26450503.

Pohler KG, Green JA, Moley LA, Doran KM, Graff HB, Peres RFG, Vasconcelos JLM, Smith MF. The effect of embryonic size and sire on circulating concentrations of bovine pregnancy associated glycoproteins in beef cattle. In: Proceedings of the International Symposium on Reproduction in Domestic Ruminants; 2014; Obihiro, Hokkaido, Japan. Ashby-de-la-Zouch, England: Context Products; 2014. p. 563.

Pohler KG, Green JA, Moley LA, Gunewardena S, Hung W-T, Payton RR, Hong X, Christenson LK, Geary TW, Smith MF. Circulating microRNA as candidates for early embryonic viability in cattle. Mol Reprod Dev. 2017;84(8):731-43. http://dx.doi.org/10.1002/mrd.22856. PMid:28643872.

Pohler KG, Pereira MHC, Lopes FR, Lawrence JC, Keisler DH, Smith MF, Vasconcelos JLM, Green JA. Circulating concentrations of bovine pregnancy-associated glycoproteins and late embryonic mortality in lactating dairy herds. J Dairy Sci. 2016a;99(2):1584-94. http://dx.doi.org/10.3168/jds.2015-10192. PMid:26709163.

Pohler KG, Peres RFG, Green JA, Graff H, Martins T, Vasconcelos JLM, Smith MF. Use of bovine pregnancy-associated glycoproteins to predict late embryonic mortality in postpartum Nelore beef cows. Theriogenology. 2016b;85(9):1652-9. http://dx.doi.org/10.1016/j.theriogenology.2016.01.026. PMid:26928645.

Pope W. Uterine asynchrony: a cause of embryonic loss. Biol Reprod. 1988;39(5):999-1003. http://dx.doi.org/10.1095/biolreprod39.5.999. PMid:3064819.

Pugliesi G, de Melo GD, Ataíde GA Jr, Pellegrino CAG, Silva JB, Rocha CC, Motta IG, Vasconcelos JLM, Binelli M. Use of Doppler ultrasonography in embryo transfer programs: feasibility and field results. Animal Reproduction (AR). 2018;15(3):239-46. http://dx.doi.org/10.21451/1984-3143-AR2018-0059.

Pugliesi G, Miagawa BT, Paiva YN, França MR, Silva LA, Binelli M. Conceptus-induced changes in the gene expression of blood immune cells and the ultrasound-accessed luteal function in beef cattle: how early can we detect pregnancy? Biol Reprod. 2014;91(4):95. http://dx.doi.org/10.1095/biolreprod.114.121525. PMid:25210129.

Pursley JR, Silcox RW, Wiltbank MC. Effect of time of artificial insemination on pregnancy rates, calving rates, pregnancy loss, and gender ratio after synchronization of ovulation in lactating dairy cows. J Dairy Sci. 1998;81(8):2139-44. http://dx.doi.org/10.3168/jds.S0022-0302(98)75790-X. PMid:9749378.

Reese ST, Franco GA, Poole RK, Hood R, Fernadez Montero L, Oliveira RV Fo, Cooke RF, Pohler KG. Pregnancy loss in beef cattle: a meta-analysis. Anim Reprod Sci. 2020;212:106251. http://dx.doi.org/10.1016/j.anireprosci.2019.106251. PMid:31864492.

Reese S, Geary T, Franco G, Moraes J, Spencer T, Pohler K. Pregnancy associated glycoproteins (PAGs) and pregnancy loss in high vs sub fertility heifers. Theriogenology. 2019;135:7-12. http://dx.doi.org/10.1016/j.theriogenology.2019.05.026. PMid:31185426.

Reese ST, Pereira MHC, Edwards JL, Vasconcelos JLM, Pohler KG. Pregnancy diagnosis in cattle using pregnancy associated glycoprotein concentration in circulation at day 24 of gestation. Theriogenology. 2018;106:178-85. http://dx.doi.org/10.1016/j.theriogenology.2017.10.020. PMid:29073542.

Reid G, Kirschner MB, van Zandwijk N. Circulating microRNAs: association with disease and potential use as biomarkers. Crit Rev Oncol Hematol. 2011;80(2):193-208. http://dx.doi.org/10.1016/j.critrevonc.2010.11.004. PMid:21145252.

Reliszko ZP, Gajewski Z, Kaczmarek MM. Signs of embryo-maternal communication: miRNAs in the maternal serum of pregnant pigs. Reproduction. 2017;154(3):217-28. http://dx.doi.org/10.1530/REP-17-0224. PMid:28592665.

Richardson B, Hill S, Stevenson J, Djira J, Perry G. Meta-analysis of the effect of estrus expression before fixed-time AI on conception rates in beef cattle. J Anim Sci. 2014;92:701.

Roberts SJ. Veterinary obstetrics and genital diseases. Woodstock, Vt: S.J. Roberts; 1956.

Roche J, Bolandl M, McGeady T. Reproductive wastage following artificial insemination of heifers. Vet Rec. 1981;109(18):401-4. http://dx.doi.org/10.1136/vr.109.18.401. PMid:7340073.

Romano JE, Larson JE. Accuracy of pregnancy specific protein-B test for early pregnancy diagnosis in dairy cattle. Theriogenology. 2010;74(6):932-9. http://dx.doi.org/10.1016/j.theriogenology.2010.04.018. PMid:20580072.

Romano JE, Thompson JA, Forrest DW, Westhusin ME, Tomaszweski MA, Kraemer DC. Early pregnancy diagnosis by transrectal ultrasonography in dairy cattle. Theriogenology. 2006;66(4):1034-41. http://dx.doi.org/10.1016/j.theriogenology.2006.02.044. PMid:16584765.

Ruhmann B, Giller K, Hankele A-K, Ulbrich S, Schmicke M. Interferon-τ induced gene expression in bovine hepatocytes during early pregnancy. Theriogenology. 2017;104:198-204. http://dx.doi.org/10.1016/j.theriogenology.2017.07.051. PMid:28888122.

Russo F, Scoyni F, Fatica A, Pellegrini M, Ferro A, Pulvirenti A, Giugno R. Circulating noncoding RNAs as clinical biomarkers. In: García-Giménez JL, editor. Epigenetic biomarkers and diagnostics. San Diego: Elsevier; 2016. p. 239-58. http://dx.doi.org/10.1016/B978-0-12-801899-6.00012-7.

Santos J, Thatcher W, Chebel R, Cerri R, Galvão K. The effect of embryonic death rates in cattle on the efficacy of estrus synchronization programs. Anim Reprod Sci. 2004;82-83:513-35. http://dx.doi.org/10.1016/j.anireprosci.2004.04.015. PMid:15271477.

Sasser R, Ruder C. Detection of early pregnancy in domestic ruminants. J Reprod Fertil Suppl. 1987;34:261-71. PMid:3305923.

Schanzenbach CI, Kirchner B, Ulbrich SE, Pfaffl MW. Can milk cell or skim milk miRNAs be used as biomarkers for early pregnancy detection in cattle? PLoS One. 2017;12(2):e0172220. http://dx.doi.org/10.1371/journal.pone.0172220. PMid:28234939.

Schlafer D, Fisher P, Davies C. The bovine placenta before and after birth: placental development and function in health and disease. Anim Reprod Sci. 2000;60-61:145-60. http://dx.doi.org/10.1016/S0378-4320(00)00132-9. PMid:10844191.

Schoggins JW. Interferon-stimulated genes: roles in viral pathogenesis. Curr Opin Virol. 2014;6:40-6. http://dx.doi.org/10.1016/j.coviro.2014.03.006. PMid:24713352.

Scully S, Evans A, Carter F, Duffy P, Lonergan P, Crowe M. Ultrasound monitoring of blood flow and echotexture of the corpus luteum and uterus during early pregnancy of beef heifers. Theriogenology. 2015;83(3):449-58. http://dx.doi.org/10.1016/j.theriogenology.2014.10.009. PMid:25459026.

Shaw AE, Hughes J, Gu Q, Behdenna A, Singer JB, Dennis T, Orton RJ, Varela M, Gifford RJ, Wilson SJ, Palmarini M. Fundamental properties of the mammalian innate immune system revealed by multispecies comparison of type I interferon responses. PLoS Biol. 2017;15(12):e2004086. http://dx.doi.org/10.1371/journal.pbio.2004086. PMid:29253856.

Sheikh AA, Hooda O, Kalyan A, Kamboj A, Mohammed S, Alhussien M, Reddi S, Shimray PG, Rautela A, Pandita S, Kapila S, De S, Dang AK. Interferon-tau stimulated gene expression: A proxy to predict embryonic mortality in dairy cows. Theriogenology. 2018;120:61-7. http://dx.doi.org/10.1016/j.theriogenology.2018.07.028. PMid:30096617.

Shirasuna K, Matsumoto H, Matsuyama S, Kimura K, Bollwein H, Miyamoto A. Possible role of IFNT on the bovine corpus luteum and neutrophils during the early pregnancy. Reproduction. 2015;150(3):217. http://dx.doi.org/10.1530/REP-15-0085.

Shirozu T, Iwano H, Ogiso T, Suzuki T, Balboula AZ, Bai H, Kawahara M, Kimura K, Takahashi H, Rulan B, Kim SW, Yanagawa Y, Nagano M, Imakawa K, Takahashi M. Estrous cycle stage-dependent manner of type I interferon-stimulated genes induction in the bovine endometrium. J Reprod Dev. 2017;63(3):2016-176. http://dx.doi.org/10.1262/jrd.2016-176. PMid:28239027.

Silva E, Sterry R, Kolb D, Mathialagan N, McGrath M, Ballam J, Fricke P. Accuracy of a pregnancy-associated glycoprotein ELISA to determine pregnancy status of lactating dairy cows twenty-seven days after timed artificial insemination. J Dairy Sci. 2007;90(10):4612-22. http://dx.doi.org/10.3168/jds.2007-0276. PMid:17881682.

Siqueira L, Areas V, Ghetti A, Fonseca J, Palhao M, Fernandes C, Viana J. Color Doppler flow imaging for the early detection of nonpregnant cattle at 20 days after timed artificial insemination. J Dairy Sci. 2013;96(10):6461-72. http://dx.doi.org/10.3168/jds.2013-6814. PMid:23958005.

Sousa N, Ayad A, Beckers J, Gajewski Z. Pregnancy-associated glycoproteins (PAG) as pregnancy markers in the ruminants. J Physiol Pharmacol. 2006;57(Suppl. 8):153-71. PMid:17242480.

Starbuck MJ, Dailey RA, Inskeep EK. Factors affecting retention of early pregnancy in dairy cattle. Anim Reprod Sci. 2004;84(1):27-39. http://dx.doi.org/10.1016/j.anireprosci.2003.12.009. PMid:15302385.

Stevenson J, Dalton J, Ott T, Racicot K, Chebel R. Correlation between reproductive status and steady-state messenger ribonucleic acid levels of the resistance gene, MX2, in peripheral blood leukocytes of dairy heifers. J Anim Sci. 2007;85(9):2163-72. http://dx.doi.org/10.2527/jas.2007-0014. PMid:17431047.

Stronge A, Sreenan J, Diskin M, Mee J, Kenny D, Morris D. Post-insemination milk progesterone concentration and embryo survival in dairy cows. Theriogenology. 2005;64(5):1212-24. http://dx.doi.org/10.1016/j.theriogenology.2005.02.007. PMid:16125563.

Surani M, Barton S, Norris M. Influence of parental chromosomes on spatial specificity in androgenetic↔ parthenogenetic chimaeras in the mouse. Nature. 1987;326(6111):395-7. http://dx.doi.org/10.1038/326395a0. PMid:3561479.

Szenci O, Humblot P, Beckers J, Sasser G, Sulon J, Baltusen R, Varga J, Bajcsy CÁ, Taverne M. Plasma profiles of progesterone and conceptus proteins in cows with spontaneous embryonic/fetal mortality as diagnosed by ultrasonography. Vet J. 2000;159(3):287-90. http://dx.doi.org/10.1053/tvjl.1999.0399. PMid:10775475.

Tabet F, Vickers KC, Torres LFC, Wiese CB, Shoucri BM, Lambert G, Catherinet C, Prado-Lourenco L, Levin MG, Thacker S. HDL-transferred microRNA-223 regulates ICAM-1 expression in endothelial cells. Nat Commun. 2014;5(1):1-14. http://dx.doi.org/10.1038/ncomms4292. PMid:24576947.

Thompson I, Cerri R, Kim I, Green J, Santos J, Thatcher W. Effects of resynchronization programs on pregnancy per artificial insemination, progesterone, and pregnancy-associated glycoproteins in plasma of lactating dairy cows. J Dairy Sci. 2010;93(9):4006-18. http://dx.doi.org/10.3168/jds.2009-2941. PMid:20723675.

Turchinovich A, Weiz L, Burwinkel B. Extracellular miRNAs: the mystery of their origin and function. Trends Biochem Sci. 2012;37(11):460-5. http://dx.doi.org/10.1016/j.tibs.2012.08.003. PMid:22944280.

Turchinovich A, Weiz L, Langheinz A, Burwinkel B. Characterization of extracellular circulating microRNA. Nucleic Acids Res. 2011;39(16):7223-33. http://dx.doi.org/10.1093/nar/gkr254. PMid:21609964.

United States Department of Agriculture – USDA. NAHMS dairy 2007 part IV: reference of dairy cattle health and management practices in the United States. Fort Collins, CO: USDA–APHIS–VS–CEAH–NAHMS; 2009. p. 33-43.

United States Department of Agriculture – USDA. NAHMS Beef 2007-2008. Fort Collins, CO: USDA–APHIS–VS–CEAH–NAHMS; 2010.

Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654-9. http://dx.doi.org/10.1038/ncb1596. PMid:17486113.

Velu VK, Ramesh R, Srinivasan A. Circulating microRNAs as biomarkers in health and disease. J Clin Diagn Res. 2012;6(10):1791-5. http://dx.doi.org/10.7860/JCDR/2012/4901.2653. PMid:23373057.

Wallace RM, Pohler KG, Smith MF, Green JA. Placental PAGs: gene origins, expression patterns, and use as markers of pregnancy. Reproduction. 2015;149(3):R115-26. http://dx.doi.org/10.1530/REP-14-0485. PMid:25661256.

Wang K, Zhang S, Weber J, Baxter D, Galas DJ. Export of microRNAs and microRNA-protective protein by mammalian cells. Nucleic Acids Res. 2010;38(20):7248. http://dx.doi.org/10.1093/nar/gkq601. PMid:20615901.

Wiltbank MC, Baez GM, Garcia-Guerra A, Toledo MZ, Monteiro PL, Melo LF, Ochoa JC, Santos JE, Sartori R. Pivotal periods for pregnancy loss during the first trimester of gestation in lactating dairy cows. Theriogenology. 2016;86(1):239-53. http://dx.doi.org/10.1016/j.theriogenology.2016.04.037. PMid:27238438.

Wooding F, Wathes DC. Binucleate cell migration in the bovine placentome. J Reprod Fertil. 1980;59(2):425-30. http://dx.doi.org/10.1530/jrf.0.0590425. PMid:7431300.

Wooding F. Structure and function of placental binucleate (giant) cells. Bibl Anat. 1982;22(22):134-9. PMid:7126144.

Yankey S, Hicks B, Carnahan K, Assiri A, Sinor S, Kodali K, Stellflug J, Ott T. Expression of the antiviral protein Mx in peripheral blood mononuclear cells of pregnant and bred, non-pregnant ewes. J Endocrinol. 2001;170(2):R7-11. http://dx.doi.org/10.1677/joe.0.170r007. PMid:11479146.

Yoshino H, Toji N, Sasaki K, Koshi K, Yamagishi N, Takahashi T, Ishiguro-Oonuma T, Matsuda H, Yamanouchi T, Hashiyada Y, Imai K, Izaike Y, Kizaki K, Hashizume K. A predictive threshold value for the diagnosis of early pregnancy in cows using interferon-stimulated genes in granulocytes. Theriogenology. 2018;107:188-93. http://dx.doi.org/10.1016/j.theriogenology.2017.11.014. PMid:29172175.

Young C, Schrick F, Pohler K, Saxton A, Di Croce F, Roper D, Wilkerson J, Edwards J. A reproductive tract scoring system to manage fertility in lactating dairy cows. J Dairy Sci. 2017;100(7):5922-7. http://dx.doi.org/10.3168/jds.2016-12288. PMid:28478009.

Zernecke A, Bidzhekov K, Noels H, Shagdarsuren E, Gan L, Denecke B, Hristov M, Köppel T, Jahantigh MN, Lutgens E, Wang S, Olson EN, Schober A, Weber C. Delivery of microRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci Signal. 2009;2(100):ra81-81. http://dx.doi.org/10.1126/scisignal.2000610. PMid:19996457.


Submitted date:
05/29/2020

Accepted date:
07/29/2020

5f6b99e40e88259e1a9fefc6 animreprod Articles
Links & Downloads

Anim Reprod

Share this page
Page Sections