Cynthia Dela Cruz

@umich.edu

University of Michigan

RESEARCH, TEACHING, or OTHER INTERESTS

Endocrinology, General Biochemistry, Genetics and Molecular Biology, Physiology

31

Scopus Publications

Scopus Publications

  • Translocator protein expression and localization in human endometrium and endometriosis: A potential target for a noninvasive diagnosis?
    Maíra Casalechi, Cynthia Dela Cruz, Wiviane A. Assis, Millene Vieira‐Lopes, Felipe Eduardo F. Lopes, Antônio M.C. Francisco, and Fernando M. Reis

    Wiley
    AbstractThe limitations of current imaging methods to detect small or superficial endometriotic lesions prompt the search for new molecular targets. TSPO is an 18 KDa protein located in the outer mitochondrial membrane, which can be traced by positron emission tomography (PET) using specific ligands. TSPO is located mostly in neurons and inflammatory sites outside the brain. We hypothesized that it might also be expressed in the human endometrium and endometrial‐like tissue, being a target for molecular imaging of endometriosis. This prospective cross‐sectional study included 28 women with endometriosis and 11 endometriosis‐free controls. Endometriotic lesions (n = 49) and normal peritoneum (n = 13) from endometriosis patients were obtained during laparoscopy, while samples of eutopic endometrium from patients with endometriosis (n = 28) and from control women (n = 11) were collected in the operating room using a flexible device. TSPO mRNA expression was evaluated by quantitative reverse‐transcription real‐time PCR while protein expression was evaluated by immunohistochemistry with a monoclonal antibody antihuman TSPO. TSPO mRNA expression was detected in an invariable fashion in all tissue types evaluated; however, TSPO protein was found to be more abundant in the glandular epithelium than in the stroma, both in the endometrium and in the endometriotic lesions. Interestingly, hormone therapies did not alter the expression of TSPO, and its presence was mostly negative in tissues adjacent to endometriotic implants. As a proof of concept, the protein expression pattern of TSPO in endometriotic tissue and along the adjacent areas suggests that TSPO‐based molecular imaging might be used for noninvasive endometriosis detection.

  • In vitro fertilization outcomes in a mouse model of gender-affirming hormone therapy in transmasculine youth
    Cynthia Dela Cruz, Abigail Wandoff, Margaret Brunette, Vasantha Padmanabhan, Ariella Shikanov, and Molly B. Moravek

    Elsevier BV

  • Impaired in vitro fertilization outcomes following testosterone treatment improve with washout in a mouse model of gender-affirming hormone treatment
    Amanda R. Schwartz, Min Xu, Nicholas C. Henderson, Cynthia Dela Cruz, Daniel Pfau, Vasantha Padmanabhan, Ariella Shikanov, and Molly B. Moravek

    Elsevier BV

  • Presence of ovarian stromal aberrations after cessation of testosterone therapy in a transgender mouse model†
    Hadrian M Kinnear, Prianka H Hashim, Cynthia Dela Cruz, Faith L Chang, Gillian Rubenstein, Likitha Nimmagadda, Venkateswaran Ramamoorthi Elangovan, Andrea Jones, Margaret A Brunette, D Ford Hannum,et al.

    Oxford University Press (OUP)
    Abstract Some transmasculine individuals may be interested in pausing gender-affirming testosterone therapy and carrying a pregnancy. The ovarian impact of taking and pausing testosterone is not completely understood. The objective of this study was to utilize a mouse model mimicking transmasculine testosterone therapy to characterize the ovarian dynamics following testosterone cessation. We injected postpubertal 9–10-week-old female C57BL/6N mice once weekly with 0.9 mg of testosterone enanthate or a vehicle control for 6 weeks. All testosterone-treated mice stopped cycling and demonstrated persistent diestrus within 1 week of starting testosterone, while control mice cycled regularly. After 6 weeks of testosterone therapy, one group of testosterone-treated mice and age-matched vehicle-treated diestrus controls were sacrificed. Another group of testosterone-treated mice were maintained after stopping testosterone therapy and were sacrificed in diestrus four cycles after the resumption of cyclicity along with age-matched vehicle-treated controls. Ovarian histological analysis revealed stromal changes with clusters of large round cells in the post testosterone group as compared to both age-matched controls and mice at 6 weeks on testosterone. These clusters exhibited periodic acid–Schiff staining, which has been previously reported in multinucleated macrophages in aging mouse ovaries. Notably, many of these cells also demonstrated positive staining for macrophage markers CD68 and CD11b. Ovarian ribonucleic acid-sequencing found upregulation of immune pathways post testosterone as compared to age-matched controls and ovaries at 6 weeks on testosterone. Although functional significance remains unknown, further attention to the ovarian stroma may be relevant for transmasculine people interested in pausing testosterone to carry a pregnancy.

  • A mouse model mimicking gender-affirming treatment with pubertal suppression followed by testosterone in transmasculine youth
    Cynthia Dela Cruz, Hadrian M Kinnear, Prianka H Hashim, Abigail Wandoff, Likitha Nimmagadda, Faith L Chang, Vasantha Padmanabhan, Ariella Shikanov, and Molly B Moravek

    Oxford University Press (OUP)
    Abstract STUDY QUESTION Can mice serve as a translational model to examine the reproductive consequences of pubertal suppression with GnRH agonist (GnRHa) followed by testosterone (T) administration, a typical therapy in peripubertal transmasculine youth? SUMMARY ANSWER An implanted depot with 3.6 mg of GnRHa followed by T enanthate at 0.45 mg weekly can be used in peripubertal female mice for investigating the impact of gender-affirming hormone therapy in transmasculine youth. WHAT IS KNOWN ALREADY There is limited knowledge available in transgender medicine to provide evidence-based fertility care, with the current guidelines being based on the assumption of fertility loss. We recently successfully developed a mouse model to investigate the reproductive consequences of T therapy given to transgender men. On the other hand, to our knowledge, there is no mouse model to assess the reproductive outcomes in peripubertal transmasculine youth. STUDY DESIGN, SIZE, DURATION A total of 80 C57BL/6N female mice were used in this study, with n = 7 mice in each experimental group. PARTICIPANTS/MATERIALS, SETTING, METHODS We first assessed the effectiveness of GnRHa in arresting pubertal development in the female mice. In this experiment, 26-day-old female mice were subcutaneously implanted with a GnRHa (3.6 mg) depot. Controls underwent a sham surgery. Animals were euthanized at 3, 9, 21 and 28 days after the day of surgery. In the second experiment, we induced a transmasculine youth mouse model. C57BL/6N female mice were subcutaneously implanted with a 3.6 mg GnRHa depot on postnatal day 26 for 21 days and this was followed by weekly injections of 0.45 mg T enanthate for 6 weeks. The control for the GnRH treatment was sham surgery and the control for T treatment was sesame oil vehicle injections. Animals were sacrificed 0.5 weeks after the last injection. The data collected included the day of the vaginal opening and first estrus, daily vaginal cytology, weekly and terminal reproductive hormones levels, body/organ weights, ovarian follicular distribution and corpora lutea (CL) counts. MAIN RESULTS AND THE ROLE OF CHANCE GnRHa implanted animals remained in persistent diestrus and had reduced levels of FSH (P = 0.0013), LH (P = 0.0082) and estradiol (P = 0.0155), decreased uterine (P < 0.0001) and ovarian weights (P = 0.0002), and a lack of CL at 21 days after GnRHa implantation. T-only and GnRHa+T-treated animals were acyclic throughout the treatment period, had sustained elevated levels of T, suppressed LH levels (P < 0.0001), and an absence of CL compared to controls (P < 0.0001). Paired ovarian weights were reduced in the T-only and GnRHa+T groups compared with the control and GnRHa-only groups. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Although it is an appropriate tool to provide relevant findings, precaution is needed to extrapolate mouse model results to mirror human reproductive physiology. WIDER IMPLICATIONS OF THE FINDINGS To our knowledge, this study describes the first mouse model mimicking gender-affirming hormone therapy in peripubertal transmasculine youth. This model provides a tool for researchers studying the effects of GnRHa-T therapy on other aspects of reproduction, other organ systems and transgenerational effects. The model is supported by GnRHa suppressing puberty and maintaining acyclicity during T treatment, lower LH levels and absence of CL. The results also suggest GnRHa+T therapy in peripubertal female mice does not affect ovarian reserve, since the number of primordial follicles was not affected by treatment. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the Michigan Institute for Clinical and Health Research grants KL2 TR 002241 and UL1 TR 002240 (C.D.C.); National Institutes of Health grants F30-HD100163 and T32-HD079342 (H.M.K.); University of Michigan Office of Research funding U058227 (A.S.); American Society for Reproductive Medicine/Society for Reproductive Endocrinology and Infertility grant (M.B.M.); and National Institutes of Health R01-HD098233 (M.B.M.). The University of Virginia Center for Research in Reproduction Ligand Assay and Analysis Core Facility was supported by the Eunice Kennedy Shriver NICHD/NIH grants P50-HD028934 and R24-HD102061. The authors declare that they have no competing interests.

  • A Mouse Model to Investigate the Impact of Gender Affirming Hormone Therapy with Estradiol on Reproduction
    Daniel R. Pfau, Amanda R. Schwartz, Cynthia Dela Cruz, Vasantha Padmanabhan, Molly B. Moravek, and Ariella Shikanov

    Wiley
    AbstractGender‐affirming hormone therapy (GAHT) can help transgender and/or gender diverse (TGD) individuals achieve emobidment goals that align with their transition needs. Clinical evidence from estradiol (E)‐GAHT patients indicate widespread changes in tissues sensitive to E and testosterone (T), particularly in the reproductive system. Notably, E‐GAHTs effects on hormones and reproduction vary greatly between patients. With the goal of informing clinical research and practice for TGD individuals taking E, this study examines intact male mice implanted with capsules containing one of three different E doses (low 1.25 mg; mid 2.5 mg; high 5 mg), or a blank control capsule. All E‐GAHT doses suppress T and follicle stimulating hormone levels while elevating E levels. Only the high E‐GAHT dose significantly supresses luteinizing hormone levels. All E‐GAHT doses affect epididymis tubule size similarly while seminiferous tubule morphology and bladder weight changes are dose‐dependent. E‐GAHT does not alter the presence of mature sperm, though E‐exposed sperm have altered motility. These data represent the first evidence that mouse models offer an effective tool to understand E‐GAHTs impact on reproductive health and the dose‐dependent effects of this model permit examinations of diverse patient outcomes.

  • Altered Endometrial Expression of a-Inhibin Subunit and Its Co-Receptor Betaglycan in Infertile Women with Endometriosis
    Cynthia Dela Cruz, Quesia T. Vilamil, Maíra Casalechi, Carolina P. Rezende, Wiviane A. Assis, Helen L. Del Puerto, Maurício Simões Abrão, and Fernando M. Reis

    S. Karger AG
    <b><i>Background:</i></b> Inhibins and their co-receptor betaglycan are members of the transforming growth factor β superfamily, a group of signaling molecules that control the differentiation of human endometrium in the secretory phase of the menstrual cycle. <b><i>Objective:</i></b> Since endometriosis is associated with endometrial dysfunction and infertility, this study aimed at evaluating the expression of α-inhibin and betaglycan mRNA and proteins in endometrial samples of infertile women with and without endometriosis. <b><i>Design:</i></b> This was a cross-sectional study. <b><i>Participants/Materials:</i></b> Endometrial samples of women with (<i>n</i> = 17) and without (<i>n</i> = 22) endometriosis were subdivided according to the menstrual cycle phase into proliferative and secretory. <b><i>Setting:</i></b> University hospital. <b><i>Methods:</i></b> We used real-time RT-PCR to quantify mRNA levels and immunohistochemistry to localize the proteins. <b><i>Results:</i></b> α-inhibin mRNA levels were significantly increased in the secretory phase (<i>p</i> < 0.01 vs. proliferative phase) only among women with endometriosis. Conversely, betaglycan mRNA levels were downregulated in the secretory endometrium of controls (<i>p</i> < 0.01 vs. proliferative) but failed to change between cycle phases of patients with endometriosis. Both proteins were present in the glandular epithelium and stroma in the endometrium of women with and without endometriosis. Immunostaining analysis showed that while α-inhibin protein expression did not vary significantly, the intensity of betaglycan immunostaining decreased in the secretory phase in the control group (<i>p</i> = 0.038 vs. proliferative phase) but not in the endometriosis group. <b><i>Limitations:</i></b> We cannot determine whether endometriosis causes the abnormal expression of α-inhibin and betaglycan in the eutopic endometrium or if this alteration already existed before the establishment of endometriotic lesions. <b><i>Conclusion:</i></b> Our findings suggest an abnormally increased expression of α-inhibin mRNA (not protein) and betaglycan (mRNA and protein) in the secretory-phase endometrium of women with endometriosis.

  • Seminal protein levels assessed with dipstick test correlate with sperm recovery after cryopreservation
    Séphora C. Queiroz, Cynthia Dela Cruz, Maíra Casalechi, Simone F. Nery, and Fernando M. Reis

    Informa UK Limited
    Abstract The process of freezing/thawing causes structural and functional damage to sperm samples, which can be mitigated by seminal plasma proteins. This study investigated the proposition that seminal protein measurements using a urinary dipstick prior to freezing could help predict the post-thaw recovery of live spermatozoa. This was a prospective study including 149 men undergoing semen analysis due to male and/or female infertility. The seminal samples were analysed according to World Health Organisation standards and protein concentrations were measured using a commercially available urinary dipstick following quantitative validation. The median live sperm recovery rates were 79%, 81% and 94%, respectively, in samples with protein concentrations of ≤1.0 g/L (+/++), 3.0 g/L (+++) and ≥20.0 g/L (++++) measured in fresh specimen dipstick analysis (p < 0.05) indicating that the probability of recovering at least 50% of frozen spermatozoa increased progressively with higher protein concentrations in the fresh sample (chi-square for linear association = 7.17. p = 0.007). In conclusion, fresh seminal protein concentration levels assessed with a dipstick test correlate with the proportion of live spermatozoa recovered from cryopreserved samples. This simple, low-cost test may add prognostic information to baseline semen analysis prior to sperm banking.

  • Immunolocalization of stem/progenitor cell biomarkers Oct-4, C-kit and Musashi-1 in endometriotic lesions
    Flavia R. Oliveira, Maíra Casalechi, Márcia M. Carneiro, Ivete de Ávila, Cynthia Dela Cruz, Helen L. Del Puerto, Aroldo F. Camargos, Maurício S. Abrão, and Fernando M. Reis

    Molecular Biology Reports Springer Science and Business Media LLC
    BACKGROUND Human endometrium harbors stem/progenitor cells (SPCs) that may contribute to the establishment of endometriosis when seeded outside the uterus. Oct-4, C-kit and Musashi-1 are some of the many proteins used to characterize SPCs, but their association with endometriosis is uncertain. OBJECTIVE AND DESIGN In this study, specimens of normal endometrium (n = 12), eutopic endometrium from women with endometriosis (n = 9), superficial peritoneal endometriosis (SUP, n = 12) and deep endometriosis (DE, n = 13) lesions were evaluated for localization and intensity of immunostaining for Oct-4, C-kit and Musashi-1. RESULTS The three markers were abundantly expressed in normal endometrium, eutopic endometrium from endometriosis patients, SUP and DE specimens. Oct-4 and C-kit expression did not vary across groups as regards intensity or frequency. C-kit staining signal was seldom detected in vascular endothelium of normal or eutopic endometrium from endometriosis patients; however, it was positive in 67% of the SUP lesions and in 25% of the DE lesions (p = 0.042). Musashi-1 was expressed in some endometriotic glands as cell clusters, but its signal was similar between the four types of tissue (p = 0.971) CONCLUSION: The wide distribution of Oct-4, C-kit and Musashi-1 in endometria of patients with and without endometriosis and in SUP and DE endometriotic lesions suggests that these markers are not suitable for the in situ characterization of endometrial SPCs and should not be taken as surrogates for the study of SPCs in the pathogenesis of endometriosis.

  • Reversibility of testosterone-induced acyclicity after testosterone cessation in a transgender mouse model
    Hadrian M. Kinnear, Prianka H. Hashim, Cynthia Dela Cruz, Gillian Rubenstein, Faith L. Chang, Likitha Nimmagadda, Margaret A. Brunette, Vasantha Padmanabhan, Ariella Shikanov, and Molly B. Moravek

    Elsevier BV

  • Conditional Fgfr1 Deletion in GnRH Neurons Leads to Minor Disruptions in the Reproductive Axis of Male and Female Mice
    Cynthia Dela Cruz, Cassandra A. Horton, Kelsey N. Sanders, Nathan D. Andersen, and Pei-San Tsai

    Frontiers Media SA
    In humans and mice, inactivating mutations in fibroblast growth factor receptor 1 (Fgfr1) lead to gonadotropin-releasing hormone (GnRH) deficiency and a host of downstream reproductive disorders. It was unclear if Fgfr1 signaling directly upon GnRH neurons critically drove the establishment of a functional GnRH system. To answer this question, we generated a mouse model with a conditional deletion of Fgfr1 in GnRH neurons using the Cre/loxP approach. These mice, called Fgfr1cKO mice, were examined along with control mice for their pubertal onset and a host of reproductive axis functions. Our results showed that Fgfr1cKO mice harbored no detectable defects in the GnRH system and pubertal onset, suffered only subtle changes in the pituitary function, but exhibited significantly disrupted testicular and ovarian morphology at 25 days of age, indicating impaired gametogenesis at a young age. However, these disruptions were transient and became undetectable in older mice. Our results suggest that Fgfr1 signaling directly on GnRH neurons supports, to some extent, the reproductive axis function in the period leading to the early phase of puberty, but is not critically required for pubertal onset or reproductive maintenance in sexually mature animals.

  • C-type natriuretic peptide signaling in human follicular environment and its relation with oocyte maturation
    Maíra Casalechi, Júlia A. Dias, Lorena V. Pinto, Verônica N. Lobach, Maria T. Pereira, Ines K. Cavallo, Adelina M. Reis, Cynthia Dela Cruz, and Fernando M. Reis

    Elsevier BV

  • Mindfulness-based program for stress reduction in infertile women: Randomized controlled trial
    Simone F. Nery, Sara P.C. Paiva, Érica L. Vieira, Andressa B. Barbosa, Edna M. Sant'Anna, Maira Casalechi, Cynthia Dela Cruz, Antônio L. Teixeira, and Fernando M. Reis

    Wiley
    Infertile women often experience chronic stress, which may have a negative impact on general well-being and may increase the burden of infertility. In this open-label, parallel, randomized controlled trial, infertile women aged 18-50 years (median 37 years) were assigned to an 8-week mindfulness-based program (MBP) or no intervention. The primary outcome was stress severity measured by the Lipp's Stress Symptoms Inventory (ISSL). Data were analyzed by modified intent-to-treat principle, which included all cases available to follow-up regardless of adherence to the intervention (62 participants from the MBP group and 37 from the control group). The median number of symptoms of chronic stress recorded in the past month decreased from six (interquartile range 2 to 9) before the MBP to two (interquartile range 1 to 4) after the intervention (p < 0.001, repeated measures analysis of variance with Time × Group interaction). Depressive symptoms also decreased after MBP, whereas general well-being improved (p < 0.01 for both outcomes). Hair cortisol and serum brain-derived neurotrophic factor (BDNF) did not change significantly between preintervention and postintervention. None of the outcomes changed significantly in the control group. MBP was effective in reducing stress and depressive symptoms while increasing general well-being in infertile women.

  • Caspase-3 gene expression in human luteinized granulosa cells is inversely correlated with the number of oocytes retrieved after controlled ovarian stimulation
    Verônica N. Lobach, Maíra Casalechi, Cynthia Dela Cruz, Maria T. Pereira, Helen L. Del Puerto, and Fernando M. Reis

    Informa UK Limited
    Abstract Granulosa cells control oocyte maturation through paracrine signalling and changes to the microenvironment around the oocyte. Apoptosis occurs as a physiological mechanism of granulosa cell renewal, but how it relates with the ovarian response to induced ovulation is still unclear. Therefore, this study evaluated apoptosis-related gene expression levels in granulosa cells of patients undergoing controlled ovarian stimulation. We enrolled prospectively 59 consecutive IVF patients referred to a tertiary academic hospital for couple infertility treatment. Luteinized granulosa cells were isolated from follicular fluid and the RNA was extracted, reverse-transcribed and the gene expression of apoptosis inducers (caspase-3, caspase-8 and bax) and inhibitor (Bcl-2) was quantified by real-time polymerase chain reaction. Caspase-3 gene expression correlated negatively with the number of pre-ovulatory follicles (Spearman’s r =  −0.308), the number of collected oocytes (r =  −0.451), the number of mature oocytes (r =  −0.526), the number of fertilized oocytes (r =  −0.439) and the number of viable embryos (r =  −0.443, all statistically significant at p < 0.02 level). No such associations were found with caspase-8, bax or bcl-2. These preliminary findings suggest that increased caspase-3 gene expression in granulosa cells is associated with a worse ovulatory response in humans.

  • Activin A in mammalian physiology
    Enrrico Bloise, Pasquapina Ciarmela, Cynthia Dela Cruz, Stefano Luisi, Felice Petraglia, and Fernando M. Reis

    American Physiological Society
    Activins are dimeric glycoproteins belonging to the transforming growth factor beta superfamily and resulting from the assembly of two beta subunits, which may also be combined with alpha subunits to form inhibins. Activins were discovered in 1986 following the isolation of inhibins from porcine follicular fluid, and were characterized as ovarian hormones that stimulate follicle stimulating hormone (FSH) release by the pituitary gland. In particular, activin A was shown to be the isoform of greater physiological importance in humans. The current understanding of activin A surpasses the reproductive system and allows its classification as a hormone, a growth factor, and a cytokine. In more than 30 yr of intense research, activin A was localized in female and male reproductive organs but also in other organs and systems as diverse as the brain, liver, lung, bone, and gut. Moreover, its roles include embryonic differentiation, trophoblast invasion of the uterine wall in early pregnancy, and fetal/neonate brain protection in hypoxic conditions. It is now recognized that activin A overexpression may be either cytostatic or mitogenic, depending on the cell type, with important implications for tumor biology. Activin A also regulates bone formation and regeneration, enhances joint inflammation in rheumatoid arthritis, and triggers pathogenic mechanisms in the respiratory system. In this 30-yr review, we analyze the evidence for physiological roles of activin A and the potential use of activin agonists and antagonists as therapeutic agents.

  • Angiotensin peptides in the non-gravid uterus: Paracrine actions beyond circulation
    Maíra Casalechi, Cynthia Dela Cruz, Luiza C. Lima, Luciana P. Maciel, Virgínia M. Pereira, and Fernando M. Reis

    Elsevier BV

  • Follistatin Expression in Human Invasive Breast Tumors: Pathologic and Clinical Associations
    Henrique L. Couto, Cynthia Dela Cruz, Marcelo A. Buzelin, Nivaldo H. Toppa, Alberto J. Wainstein, and Fernando M. Reis

    Ovid Technologies (Wolters Kluwer Health)
    Follistatin is a potent native activin antagonist that is expressed in the normal mammary gland and in different breast proliferative diseases. Despite experimental evidence that follistatin can modulate the breast cancer cell cycle, the clinical significance of follistatin expression in these tumors is unknown. The aim of this study was to correlate the intensity of follistatin expression in invasive breast cancer with some of its clinical and pathologic features, such as the disease stage and the hormonal receptor status. Paraffin blocks of tumor samples that had been fixed in buffered formalin were obtained from 154 women subjected to surgery for breast cancer between 2008 and 2012. Sections from all paraffin blocks were cut and processed together by immunohistochemistry using a commercial monoclonal antibody to human follistatin. The intensity of follistatin staining was unrelated to the menopausal status, the disease stage, the grade, progesterone receptor expression, and local or systemic recurrence. However, follistatin immunoreactivity was significantly stronger in estrogen receptor (ER)-negative tumors than in ER-positive tumors. These findings suggest that follistatin expression in invasive breast cancer is unrelated to the disease severity and the risk of recurrence, but is more intense in ER-negative tumors.

  • Deep Infiltrating Endometriosis and Endometrial Adenocarcinoma Express High Levels of Myostatin and Its Receptors Messenger RNAs
    Patrizia Carrarelli, Lucia Funghi, Pasquapina Ciarmela, Gabriele Centini, Fernando M. Reis, Cynthia Dela Cruz, Alberto Mattei, Silvia Vannuccini, and Felice Petraglia

    Springer Science and Business Media LLC
    Myostatin is a growth factor member of the transforming growth factor β superfamily, which is known to play major roles in cell proliferation and differentiation. The present study investigated the messenger RNA (mRNA) expression of myostatin and myostatin receptors (activin receptor-like kinase 4 [ALK4], transforming growth factor (TGF)-β type I receptor kinase [ALK5] and activin receptor type IIB [ActRIIB]) in endometrium of healthy women during menstrual cycle as well as in benign (endometriosis, polyps) and malignant (endometrial adenocarcinoma) conditions. Endometrial specimens were collected by hysteroscopy, whereas endometriotic lesions were collected by laparoscopy, and adenocarcinomas were sampled after hysterectomy. Total RNA was extracted from tissue homogenates, and gene expression was assessed by quantitative real-time polymerase chain reaction. Myostatin and myostatin receptors mRNAs were expressed by healthy endometrium throughout the menstrual cycle, with no differences between the proliferative and secretory phase. The highest myostatin mRNA expression was found in patients with deep infiltrating endometriosis (DIE) and in endometrial carcinoma; expression was also found in ovarian endometrioma (OMA ) and endometrial polyps. Myostatin receptors mRNA expression was higher in DIE and adenocarcinomas compared to control endometrium. The expression of ALK5 and ActRIIB in OMA was higher than in controls, whereas polyps had an increased expression of ALK5 mRNA. In conclusion, the present data showed for the first time the expression of myostatin in healthy endometrium and a higher expression in endometriosis and endometrial cancer, suggesting myostatin involvement in human endometrial physiology and related pathologies.

  • Angiotensin-(1-7) in human follicular fluid correlates with oocyte maturation
    Ines K. Cavallo, Cynthia Dela Cruz, Marilene L. Oliveira, Helen L. Del Puerto, Júlia A. Dias, Veronica N. Lobach, Maíra Casalechi, Maria G. Camargos, Adelina M. Reis, Robson A. Santos,et al.

    Oxford University Press (OUP)
    STUDY QUESTION Do angiotensin (Ang)-(1-7) levels in human ovarian follicular fluid (FF) correlate with the number and proportion of mature oocytes obtained for IVF? SUMMARY ANSWER The present study shows for the first time that Ang-(1-7) levels in human FF correlate with the proportion of mature oocytes collected upon ovarian stimulation for IVF. WHAT IS KNOWN ALREADY Ang-(1-7) is an active peptide of the renin-angiotensin system that stimulates oocyte maturation in isolated rabbit and rat ovaries. However, its role in human ovulation remains unexplored. STUDY DESIGN, SIZE, DURATION This was a prospective cohort study including 64 participants from a single IVF center. Sample size was calculated to achieve a statistical power of 80% in detecting 20% differences in the proportion of mature oocytes between groups. The participants were enrolled in the study during six consecutive months. PARTICIPANTS/MATERIALS, SETTING, METHODS Plasma samples were obtained from all subjects at Day 21 of the last menstrual cycle before starting pituitary blockade and controlled ovarian stimulation (COS). Plasma and FF samples were quickly mixed with a protease inhibitor cocktail and stored at -80°C. Ang-(1-7) was quantified in plasma and FF samples by a highly sensitive and specific radioimmunoassay, which was preceded by solid phase extraction, speed vacuum concentration and sample reconstitution in assay buffer. FF Ang-(1-7) levels were stratified into tertiles and the patients of each tertile were compared for COS/IVF outcomes using Kruskal-Wallis ANOVA. Multiple regression analysis was used to adjust correlations for potential confounders. The mRNA encoding for Mas, a receptor for Ang-(1-7), was investigated by real-time PCR in luteinized granulosa cells purified from the FF. MAIN RESULTS AND THE ROLE OF CHANCE There was a four-fold increase in plasma Ang-(1-7) after ovulation induction (median 160.9 vs 41.4 pg/ml, P < 0.0001). FF Ang-(1-7) levels were similar to (169.9 pg/ml) but did not correlate with plasma Ang-(1-7) levels (r = -0.05, P = 0.665). Patients at the highest FF Ang-(1-7) tertile had a higher proportion of mature oocytes compared to patients at the lower FF Ang-(1-7) tertile (median 100% vs 70%, P < 0.01). There was a linear correlation between FF Ang-(1-7) and the proportion of mature oocytes (r = 0.380, P < 0.01), which remained significant after adjustment for age and duration of infertility (r = 0.447, P < 0.001). The luteinized granulosa cells expressed Mas receptor mRNA, which was positively correlated to the number of mature oocytes in women with more than three mature oocytes retrieved (r = 0.42, P < 0.01). LIMITATIONS, REASONS FOR CAUTION This is an observational study, therefore, no causal relationship can be established between Ang-(1-7) and human oocyte maturation. Mas protein expression was not quantified due to limited availability of granulosa cells. WIDER IMPLICATIONS OF THE FINDINGS Since this peptide promotes oocyte maturation in other species, it deserves further investigation as a potential maturation factor to human oocytes. STUDY FUNDING AND COMPETING INTEREST(S) Research supported by Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and Fundação de Amparo à Pesquisa do Estado de Minas Gerais (FAPEMIG). The authors have nothing to disclose.

  • Urocortin and corticotrophin-releasing hormone receptor type 2 mRNA are highly expressed in deep infiltrating endometriotic lesions
    Patrizia Carrarelli, Alice Luddi, Lucia Funghi, Felice Arcuri, Frederic Batteux, Cynthia Dela Cruz, Claudia Tosti, Fernando M. Reis, Charles Chapron, and Felice Petraglia

    Elsevier BV

  • Apoptosis modulation by activin A and follistatin in human endometrial stromal cells
    Larissa M. Coutinho, Erica L. Vieira, Cynthia Dela Cruz, Maíra Casalechi, Antonio L. Teixeira, Helen L. Del Puerto, and Fernando M. Reis

    Informa UK Limited
    Abstract Activin A is a growth factor that stimulates decidualization and is abundantly expressed in endometrial proliferative disorders. Nevertheless, whether it directly affects endometrial cell survival is still unknown. This study investigated the effects of activin A on total death and apoptosis rates and on tumor necrosis factor (TNF) release by human endometrial stromal cells (HESC). We performed a controlled prospective in vitro study using primary HESC cultures obtained from healthy reproductive age women (n = 11). Cells were treated with medium alone (control) or activin A (25 ng/mL) or activin A (25 ng/mL) and its antagonist follistatin (250 ng/mL). Apoptosis and total cell death were measured by flow cytometry, while TNF concentrations in culture media were quantified by ELISA. Activin A decreased the percentage of apoptotic/dead cells from 31% to 22% (p < 0.05, paired t-test) and reduced TNF levels in culture medium by 14%, but there was no linear correlation between TNF release and apoptotic rates. Both effects of activin A were reversed by follistatin. These findings indicate that activin A promotes HESC survival, possibly by a TNF-independent pathway. This mechanism may be critical to the actions of activin A upon stromal cell growth and differentiation in physiology and disease.


  • Expression, localization and control of activin A release from human umbilical vein endothelial cells
    Lavinia E. Borges, Enrrico Bloise, Cynthia Dela Cruz, Lauretta Massai, Pasquapina Ciarmela, Rosanna Apa, Stefano Luisi, Filiberto M. Severi, Felice Petraglia, and Fernando M. Reis

    Informa UK Limited
    Abstract Activin-A is a member of the TGFβ superfamily found in maternal and umbilical cord blood throughout gestation. We investigated whether human umbilical vein endothelial cells (HUVEC) express activin-A in vivo and tested the effects of vasoactive (endothelin-1), pro-inflammatory (interferon-γ, interleukin-8) and anti-inflammatory (dexamethasone, urocortin) factors on activin-A release by isolated HUVEC in vitro. Activin βA subunit protein and mRNA were strongly localized in the endothelial cells of umbilical veins and were also detectable in scattered cells of the cord connective tissue. Dimeric activin-A was detected in the HUVEC culture medium at picomolar concentrations. Activin-A release by HUVEC decreased after cell incubation with urocortin (p < 0.01), whereas no effect was observed with interleukin-8, interferon-γ, endothelin-1 or dexamethasone. In summary, activin-A is present in the human umbilical vein endothelium in vivo and is produced and released by isolated HUVEC. Activin-A secretion is inhibited in vitro by urocortin, a neuropeptide with predominantly anti-inflammatory action.

  • The role of TGF β superfamily members in the pathophysiology of endometriosis
    Cynthia Dela Cruz and Fernando M. Reis

    Informa UK Limited
    Abstract The transforming growth factor-beta (TGFβ) superfamily comprises over 30 dimeric proteins with conserved structures, which play important roles in the control of cellular proliferation, differentiation and apoptosis. These proteins are expressed and finely regulated in human endometrium during the menstrual cycle, which is consistent with their effects on endometrial cell proliferation and tissue remodeling. This review is focused on summarizing the role of key members of the TGFβ superfamily in the pathophysiology of endometriosis. Evidence suggests that TGFβ, activins, inhibins, nodal, bone morphogenetic proteins, growth differentiation factors, and anti-Müllerian hormone are produced by endometriotic lesions and could be involved in the establishment and progression of the disease. Their receptors and signaling pathways may also be altered in the presence of endometriosis and may be potential targets to the development of therapeutic agents.

  • Expression of nodal, cripto, SMAD3, phosphorylated SMAD3, and SMAD4 in the proliferative endometrium of women with endometriosis
    Cynthia Dela Cruz, Helen L. Del Puerto, Ana Luiza L. Rocha, Inês K. Cavallo, Alessandra D. Clarizia, Felice Petraglia, and Fernando M. Reis

    Springer Science and Business Media LLC
    Background: Nodal is a growth factor of the transforming growth factor β superfamily that is expressed in high turnover tissues, such as the human endometrium, and in several malignancies. The effects of Nodal are modulated by the coreceptor Cripto and mediated by SMAD proteins. This study evaluated the gene and protein expression of Nodal, Cripto, total and phosphorylated (p) SMAD3, and SMAD4 in the proliferative endometrium of women with and without endometriosis. Method: Total RNA was isolated and complementary DNA synthesized from eutopic endometrium of women with (n = 15) and without (n = 12) endometriosis, followed by quantitative real-time polymerase chain reaction (PCR) to evaluate the gene expression of Nodal, Cripto, SMAD3, and SMAD4. Western blot was used to evaluate the protein levels of Nodal and Cripto, and immunohistochemistry was performed to localize SMAD3, pSMAD3, and SMAD4. Results: Although Nodal expression was unchanged in women with endometriosis, real-time PCR indicated lower gene expression of Cripto (fold change 0.27, P < .05) in the endometriosis group. This difference, however, was not maintained at protein expression level as assessed by Western blot. The immunostaining of total SMAD3 was reduced in the endometriosis group (P < .01), but the localization of pSMAD3 and the nuclear staining of SMAD4 were unchanged. Conclusion: These findings suggest that the Nodal signaling pathway has subtle changes in the endometrium of women with endometriosis, but this imbalance may not cause functional damage as it seems not to affect the nuclear expression of SMAD4.