Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Frank F. Bartol is active.

Publication


Featured researches published by Frank F. Bartol.


Biology of Reproduction | 2001

Developmental biology of uterine glands

C. Allison Gray; Frank F. Bartol; Becky J. Tarleton; Anne A. Wiley; Greg A. Johnson; Fuller W. Bazer; Thomas E. Spencer

Abstract All mammalian uteri contain endometrial glands that synthesize or transport and secrete substances essential for survival and development of the conceptus (embryo/fetus and associated extraembryonic membranes). In rodents, uterine secretory products of the endometrial glands are unequivocally required for establishment of uterine receptivity and conceptus implantation. Analyses of the ovine uterine gland knockout model support a primary role for endometrial glands and, by default, their secretions in peri-implantation conceptus survival and development. Uterine adenogenesis is the process whereby endometrial glands develop. In humans, this process begins in the fetus, continues postnatally, and is completed during puberty. In contrast, endometrial adenogenesis is primarily a postnatal event in sheep, pigs, and rodents. Typically, endometrial adenogenesis involves differentiation and budding of glandular epithelium from luminal epithelium, followed by invagination and extensive tubular coiling and branching morphogenesis throughout the uterine stroma to the myometrium. This process requires site-specific alterations in cell proliferation and extracellular matrix (ECM) remodeling as well as paracrine cell-cell and cell-ECM interactions that support the actions of specific hormones and growth factors. Studies of uterine development in neonatal ungulates implicate prolactin, estradiol-17β, and their receptors in mechanisms regulating endometrial adenogenesis. These same hormones appear to regulate endometrial gland morphogenesis in menstruating primates and humans during reconstruction of the functionalis from the basalis endometrium after menses. In sheep and pigs, extensive endometrial gland hyperplasia and hypertrophy occur during gestation, presumably to provide increasing histotrophic support for conceptus growth and development. In the rabbit, sheep, and pig, a servomechanism is proposed to regulate endometrial gland development and differentiated function during pregnancy that involves sequential actions of ovarian steroid hormones, pregnancy recognition signals, and lactogenic hormones from the pituitary or placenta. That disruption of uterine development during critical organizational periods can alter the functional capacity and embryotrophic potential of the adult uterus reinforces the importance of understanding the developmental biology of uterine glands. Unexplained high rates of peri-implantation embryonic loss in humans and livestock may reflect defects in endometrial gland morphogenesis due to genetic errors, epigenetic influences of endocrine disruptors, and pathological lesions.


Biology of Reproduction | 2001

Endometrial Glands Are Required for Preimplantation Conceptus Elongation and Survival

C. Allison Gray; Kristin M. Taylor; W. Shawn Ramsey; Jonathan R. Hill; Fuller W. Bazer; Frank F. Bartol; Thomas E. Spencer

Abstract Endometrial glands secrete molecules hypothesized to support conceptus growth and development. In sheep, endometrial gland morphogenesis occurs postnatally and can be epigenetically ablated by neonatal progestin exposure. The resulting stable adult uterine gland knockout (UGKO) phenotype was used here to test the hypothesis that endometrial glands are required for successful pregnancy. Mature UGKO ewes were bred repeatedly to fertile rams, but no pregnancies were detected by ultrasound on Day 25. Day 7 blastocysts from normal superovulated ewes were then transferred synchronously into Day 7 control or UGKO ewes. Ultrasonography on Days 25–65 postmating indicated that pregnancy was established in control, but not in UGKO ewes. To examine early uterine-embryo interactions, four control and eight UGKO ewes were bred to fertile rams. On Day 14, their uteri were flushed. The uterus of each control ewe contained two filamentous conceptuses of normal length. Uteri from four UGKO ewes contained no conceptus. Uteri of three UGKO ewes contained a single severely growth-retarded tubular conceptus, whereas the remaining ewe contained a single filamentous conceptus. Histological analyses of these uteri revealed that endometrial gland density was directly related to conceptus survival and developmental state. Day 14 UGKO uteri that were devoid of endometrial glands did not support normal conceptus development and contained either no conceptuses or growth-retarded tubular conceptuses. The Day 14 UGKO uterus with moderate gland development contained a filamentous conceptus. Collectively, these results demonstrate that endometrial glands and, by inference, their secretions are required for periimplantation conceptus survival and development.


Endocrinology | 1999

Discovery and characterization of endometrial epithelial messenger ribonucleic acids using the ovine uterine gland knockout model

Thomas E. Spencer; Allison G. Stagg; Margaret M. Joyce; Guido Jenster; Christopher G. Wood; Fuller W. Bazer; Anne A. Wiley; Frank F. Bartol

Prolonged exposure of the developing neonatal ovine uterus to a progestin from birth prevents uterine gland development and creates an adult endometrial phenotype characterized by the absence of glandular epithelium, the uterine gland knockout (UGKO) phenotype. This study used endometrium from normal and UGKO sheep to identify messenger RNAs (mRNAs) expressed differentially in the endometrial epithelium using the molecular techniques of mRNA differential display PCR (DD-PCR) and suppression subtractive complementary DNA (cDNA) hybridization (SSH). Sequence analyses of DD- and SSH-identified and cloned cDNAs indicated similarity of some to known mRNAs, including β-lactoglobulin, alkaline phosphatase, type B and D endogenous sheep retroviruses, gp330/megalin, matrix Gla protein, and others. Other cDNAs were not similar to any known sequences and are considered novel, although some of these match human expressed sequence tags. In situ hybridization analyses of uteri from cyclic and pregnant ewes indicated th...


Biology of Reproduction | 2000

Ovine Uterine Gland Knock-Out Model: Effects of Gland Ablation on the Estrous Cycle

C. Allison Gray; Frank F. Bartol; Kristin M. Taylor; Anne A. Wiley; W. Shawn Ramsey; Troy L. Ott; Fuller W. Bazer; Thomas E. Spencer

Abstract Ovine endometrial gland development is a postnatal event that can be inhibited epigenetically by chronic exposure of ewe lambs to a synthetic progestin from birth to puberty. As adults, these neonatally progestin-treated ewes lack endometrial glands and display a uterine gland knockout (UGKO) phenotype that is useful as a model for study of endometrial function. Here, objectives were to determine: 1) length of progestin exposure necessary from birth to produce the UGKO phenotype in ewes; 2) if UGKO ewes display normal estrous cycles; and 3) if UGKO ewes could establish and/or maintain pregnancy. Ewe lambs (n = 22) received a Norgestomet (Nor) implant at birth and every two weeks thereafter for 8 (Group I), 16 (Group II), or 32 (Groups III and IV) weeks. Control ewe lambs (n = 13) received no Nor treatment (Groups V and VI). Ewes in Groups I, II, III, and VI were hemihysterectomized (Hhx) at 16 weeks of age. After puberty, the remaining uterine horn in Hhx ewes was removed on either Day 9 or 15 of the estrous cycle (Day 0 = estrus). Histological analyses of uteri indicated that progestin exposure for 8, 16, or 32 weeks prevented endometrial adenogenesis and produced the UGKO phenotype in adult ewes. Three endometrial phenotypes were consistently observed in Nor-treated ewes: 1) no glands, 2) slight glandular invaginations into the stroma, and 3) limited numbers of cyst- or gland-like structures in the stroma. Overall patterns of uterine progesterone, estrogen, and oxytocin receptor expression were not different in uteri from adult cyclic control and UGKO ewes. However, receptor expression was variegated in the ruffled luminal epithelium of uteri from UGKO ewes. Intact UGKO ewes displayed altered estrous cycles with interestrous intervals of 17 to 43 days, and they responded to exogenous prostaglandin F2∝ (PGF) with luteolysis and behavioral estrus. During the estrous cycle, plasma concentrations of progesterone in intact control and UGKO ewes were not different during metestrus and diestrus, but levels did not decline in many UGKO ewes during late diestrus. Peak peripheral plasma concentrations of PGF metabolite, in response to an oxytocin challenge on Day 15, were threefold lower in UGKO compared to control ewes. Intact UGKO ewes bred repeatedly to intact rams did not display evidence of pregnancy based on results of ultrasound. Collectively, results indicate that 1) transient, progestin-induced disruption of ovine uterine development from birth alters both structural and functional integrity of the adult endometrium; 2) normal adult endometrial integrity, including uterine glands, is required to insure a luteolytic pattern of PGF production; and 3) the UGKO phenotype, characterized by the absence of endometrial glands and a compact, disorganized endometrial stroma, limits or inhibits the capacity of uterine tissues to support the establishment and/or maintenance of pregnancy.


Reproduction in Domestic Animals | 2008

Epigenetic Programming of Porcine Endometrial Function and the Lactocrine Hypothesis

Frank F. Bartol; Anne A. Wiley; Carol A. Bagnell

Epigenetic programs controlling development of the female reproductive tract (FRT) are influenced by the effects of naturally occurring bioactive agents on patterns of gene expression in FRT tissues during organizationally critical periods of foetal and perinatal life. Aberrations in such important cellular and molecular events, as may occur with exposure to natural or manmade steroid or peptide receptor-modulating agents, disrupt the developmental program and can change the developmental trajectory of FRT tissues, including the endometrium, with lasting consequences. In the pig, as in other mammals, maternal programming of FRT development begins pre-natally and is completed post-natally, when maternal effects on development can be communicated via signals transmitted in milk. Studies involving relaxin (RLX), a prototypic milk-borne morphoregulatory factor (MbF), serve as the basis for ongoing efforts to identify maternal programming events that affect uterine and cervical tissues in the neonatal pig. Data support the lactocrine hypothesis for delivery of MbFs to neonates as a specific consequence of nursing. Components of a maternally driven lactocrine mechanism for RLX-mediated signalling in neonatal FRT tissues, including evidence that milk-borne RLX is delivered into the neonatal circulation where it can act on RLX receptor (RXFP1)-positive neonatal tissues to affect their development, are in place in the pig. The fact that all newborn mammals drink milk extends the timeframe of maternal influence on neonatal development across many species. Thus, lactocrine transmission of milk-borne developmental signals is an element of the maternal epigenetic programming equation that deserves further study.


Molecular Human Reproduction | 2013

Uterine glands: development, function and experimental model systems

Paul S. Cooke; Thomas E. Spencer; Frank F. Bartol; Kanako Hayashi

Development of uterine glands (adenogenesis) in mammals typically begins during the early post-natal period and involves budding of nascent glands from the luminal epithelium and extensive cell proliferation in these structures as they grow into the surrounding stroma, elongate and mature. Uterine glands are essential for pregnancy, as demonstrated by the infertility that results from inhibiting the development of these glands through gene mutation or epigenetic strategies. Several genes, including forkhead box A2, beta-catenin and members of the Wnt and Hox gene families, are implicated in uterine gland development. Progestins inhibit uterine epithelial proliferation, and this has been employed as a strategy to develop a model in which progestin treatment of ewes for 8 weeks from birth produces infertile adults lacking uterine glands. More recently, mouse models have been developed in which neonatal progestin treatment was used to permanently inhibit adenogenesis and adult fertility. These studies revealed a narrow and well-defined window in which progestin treatments induced permanent infertility by impairing neonatal gland development and establishing endometrial changes that result in implantation defects. These model systems are being utilized to better understand the molecular mechanisms underlying uterine adenogenesis and endometrial function. The ability of neonatal progestin treatment in sheep and mice to produce infertility suggests that an approach of this kind may provide a contraceptive strategy with application in other species. Recent studies have defined the temporal patterns of adenogenesis in uteri of neonatal and juvenile dogs and work is underway to determine whether neonatal progestin or other steroid hormone treatments might be a viable contraceptive approach in this species.


Prostaglandins | 1986

Uterine prostaglandin and blood flow responses to estradiol-17β in cyclic cattle

J.J. Knickerbocker; W.W. Thatcher; D.B. Foster; D. Wolfenson; Frank F. Bartol; D. Caton

Normal cyclic dairy cattle (n = 7) underwent a midventral laparotomy on day 17 of the estrous cycle and were fitted, ipsilateral to the CL, with: an electromagnetic flow transducer around the uterine artery (UA; n = 5); catheters within the ovarian vein (OV; n = 7) via a uterine branch of the ovarian vein, uterine branch of the ovarian artery (UBOA; n = 5) and facial artery (FA; n = 7). On day 18, blood samples were collected at 30 min intervals for 1 h prior to injection of estradiol-17 beta (E2; 3 mg) and 12 h post-E2. Uterine blood flow (UBF) was monitored continuously and plasma samples analyzed for PGF2 alpha and PGFM. Exact locations of catheters in reproductive tracts were verified post-slaughter. Data were analyzed by method of least squares analysis of variance. Uterine blood flow (ml/min) increased above pre-E2 flow rates within 30 min post-E2 injection, peaked between 2.5 to 3.5 h and declined between 4 to 8.5 h. A small secondary rise in UBF occurred between 9 and 12 h. Regression analysis for concentrations (pg/ml) of PGF2 alpha and PGFM in the OV (i.e., [OV]-[FA]) demonstrate a similar response as PGFM concentration in the FA in that all increased at approximately 3 h, peaked between 5 and 7 h and returned to near baseline levels by 9 to 10 h post-E2. Facial artery PGFM concentrations were positively correlated with uterine production of PGF2 alpha (r = .66) and PGFM (r = .30), whereas FA PGF2 alpha concentrations were not. In three of five cows, a difference in PGF2 alpha was detected between UBOA and FA (UBOA greater than FA); supportive of a local countercurrent exchange between the uterine venous drainage and the ovarian artery.


Anatomical Record-advances in Integrative Anatomy and Evolutionary Biology | 1997

IMMUNOLOCALIZATION OF ANDROGEN RECEPTOR AND ESTROGEN RECEPTOR IN THE DEVELOPING TESTIS AND EXCURRENT DUCTS OF GOATS

Hari O. Goyal; Frank F. Bartol; Anne A. Wiley; Mohammed K. Khalil; Jiliang Chiu; Madan M. Vig

Because of the significance of androgens and estrogens in prenatal and postnatal differentiation of the testis and excurrent ducts, it is important to understand the developmental pattern of androgen receptor (AR) and estrogen receptor (ER) in these organs.


Biology of Reproduction | 2012

Brief Exposure to Progesterone During a Critical Neonatal Window Prevents Uterine Gland Formation in Mice

Paul S. Cooke; Gail C. Ekman; Jaspreet Kaur; Juanmahel Davila; Indrani C. Bagchi; Sherrie G. Clark; Philip J. Dziuk; Kanako Hayashi; Frank F. Bartol

ABSTRACT Uterine gland development (adenogenesis) in mice begins on Postnatal Day (PND) 5 and is completed in adulthood. Adenogenesis depends on estrogen receptor 1, and progesterone (P4) inhibits mitogenic effects of estrogen on uterine epithelium. This progestin-induced effect has been used to inhibit uterine gland development; progestin treatment of ewes for 8 wk from birth has produced infertile adults lacking uterine glands. The goals of the present study were to determine if a window of susceptibility to P4-mediated inhibition of uterine gland development exists in mice and whether early P4 treatment abolishes adenogenesis and fertility. Mice were injected daily with P4 (40 μg/g) or vehicle during various postnatal windows. Adenogenesis, cell proliferation, and expression of key morphoregulatory transcripts and proteins were examined in uteri at PNDs 10 and 20. Additionally, adenogenesis was assessed in isolated uterine epithelium. Treatment during PNDs 3–9, 5–9, or 3–7 abolished adenogenesis at PND 10, whereas treatments during PNDs 3–5 and 7–9 did not. Critically, mice treated during PNDs 3–9 lacked glands in adulthood, indicating that adenogenesis did not resume after this treatment. However, glands were present by PND 20 and later following treatment during PNDs 5–9 or 3–7, whereas treatment during PNDs 10–16 produced partial inhibition of adenogenesis at PND 20 and later. Epithelial proliferation at PND 10 was low following P4 treatment (PNDs 3–9) but exceeded that in controls at PND 20, indicating a rebound of epithelial proliferation following treatment. Messenger RNA for Wnt, Fzd, and Hox genes was altered by neonatal P4 treatment. All groups cycled during adulthood. Mice treated with P4 during PNDs 3–9, but not during other developmental windows, showed minimal fertility in adulthood. In summary, brief P4 treatment (7 days) during a critical neonatal window (PNDs 3–9) transiently inhibited epithelial proliferation but totally and permanently blocked adenogenesis and adult fertility. This resulted in permanent loss of uterine glands and, essentially, total infertility during adulthood. The narrow window for inhibition of adenogenesis identified here may have implications for development of this methodology as a contraceptive strategy for animals.


Journal of Animal Science | 2013

Lactation Biology Symposium: lactocrine signaling and developmental programming.

Frank F. Bartol; Anne A. Wiley; Dori J. Miller; A. J. Silva; K. E. Roberts; M. L. P. Davolt; Joseph C. Chen; Amy-Lynn Frankshun; Meredith E. Camp; Kathleen M. Rahman; J. L. Vallet; Carol A. Bagnell

Lactocrine signaling is defined as transmission of bioactive factors from mother to offspring as a consequence of nursing. Lactocrine transmission of signaling molecules may be an evolutionarily conserved process through which bioactive factors necessary for support of neonatal development are delivered postnatally. Dependence on maternal resources for development in eutherian mammals extends into neonatal life for at least that period of time when nutrition is obtained solely from first milk (i.e., colostrum). Data for the pig (Sus scrofa domesticus) provide evidence of lactocrine mediated effects on development of the female reproductive tract and other somatic tissues. Porcine uterine gland development, an estrogen receptor-alpha (ESR1)-dependent process, begins within 2 d of birth [postnatal day (PND) 0]. A lactocrine-driven, ESR1-mediated process was proposed as a regulatory mechanism governing onset of uterine gland development and endometrial maturation in the neonatal pig. Gilts maintained in a lactocrine-null state for 2 d from birth by milk-replacer feeding displayed altered patterns of endometrial gene expression and retarded uterine gland development by PND 14. In lactocrine-null gilts, inhibition of endometrial and cervical ESR1 and vascular endothelial growth factor (VEGFA) expression observed on PND 2 persisted to PND 14, even after gilts were returned to nursing on PND 2. Collectively, data support a role for lactocrine signaling in regulation of critical neonatal developmental events. Maternal lactocrine programming of postnatal development may help to insure healthy developmental outcomes. A systems biology approach will be required to define and understand mechanistic dynamics of lactocrine signaling events that may ultimately connect genotype to phenotype and establish the parameters of reproductive potential.

Collaboration


Dive into the Frank F. Bartol's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph C. Chen

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge