Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (500)

Search Parameters:
Keywords = menstrual cycle

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 650 KiB  
Review
Kisspeptin and Endometriosis—Is There a Link?
by Blazej Meczekalski, Agata Nowicka, Stefania Bochynska, Aleksandra Szczesnowicz, Gregory Bala and Anna Szeliga
J. Clin. Med. 2024, 13(24), 7683; https://doi.org/10.3390/jcm13247683 - 17 Dec 2024
Viewed by 261
Abstract
This article presents a narrative review that explores the potential link between kisspeptin—a key regulator of the hypothalamic-pituitary-gonadal axis—and the pathogenesis of endometriosis. Kisspeptin plays a significant role in regulating reproductive functions by modulating the release of gonadotropin-releasing hormone (GnRH), which in turn [...] Read more.
This article presents a narrative review that explores the potential link between kisspeptin—a key regulator of the hypothalamic-pituitary-gonadal axis—and the pathogenesis of endometriosis. Kisspeptin plays a significant role in regulating reproductive functions by modulating the release of gonadotropin-releasing hormone (GnRH), which in turn stimulates the secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Recent studies suggest that kisspeptin may also impact peripheral reproductive tissues and influence inflammatory processes involved in the development of endometriosis. Altered kisspeptin signaling has been associated with the abnormal hormonal environment observed in endometriosis, which affects menstrual cycles and ovarian function. Research indicates that women with endometriosis exhibit altered levels of kisspeptin and its receptor, KISS1R, in both eutopic and ectopic endometrial tissues, suggesting a role in disease progression, particularly in tissue invasion and lesion formation. Kisspeptin’s role in regulating matrix metalloproteinases (MMPs), enzymes essential for tissue remodeling, further supports its potential contribution to the pathophysiology of endometriosis. Moreover, kisspeptin-based therapeutic strategies are currently under investigation, with the aim of providing targeted treatments that reduce the side effects commonly associated with existing therapies. Despite promising findings, further research is needed to fully understand the mechanisms by which kisspeptin influences endometriosis. Full article
(This article belongs to the Special Issue Recent Developments in Gynecological Endocrinology)
Show Figures

Figure 1

Figure 1
<p>Kisspeptins impact on steroidogenesis through its influence on the hipothalmic-pituitary-ovarian axis. Based on [<a href="#B24-jcm-13-07683" class="html-bibr">24</a>].</p>
Full article ">Figure 2
<p>The roles of kisspeptin and KISS1R in physiological and pathological states of the endometrium of a non-pregnant women. Based on [<a href="#B49-jcm-13-07683" class="html-bibr">49</a>].</p>
Full article ">
18 pages, 2055 KiB  
Review
Menstrual Disorders in Adolescence: Diagnostic and Therapeutic Challenges
by Christiane Anthon, Marcel Steinmann, Angela Vidal and Carolin Dhakal
J. Clin. Med. 2024, 13(24), 7668; https://doi.org/10.3390/jcm13247668 - 16 Dec 2024
Viewed by 270
Abstract
Background: Adolescence is the period of life between the ages of 10 and 19. This period is essentially dominated by puberty. The first menstruation, called menarche, occurs, on average, at the age of 12–13. The period after menarche, especially the first 2 [...] Read more.
Background: Adolescence is the period of life between the ages of 10 and 19. This period is essentially dominated by puberty. The first menstruation, called menarche, occurs, on average, at the age of 12–13. The period after menarche, especially the first 2 years, is characterized by anovulatory cycles, which can be accompanied by menstrual irregularities. This review aims to describe the current status of the diagnostic and therapeutic challenges of the physiological and pathological causes of menstrual irregularities in adolescence and evaluates the benefits from interdisciplinary collaboration to ensure optimal care. Methods: A systematic literature search was conducted in the PubMed database in April 2024 using the following term: “menstrual disorder adolescence”. A total of 1724 abstracts were screened, and relevant articles from the last 10 years were included. In addition, a supplementary topic-relevant literature search of the guidelines of the European Society of Human Reproduction and Embryology (ESHRE) and the guidelines of the Arbeitsgemeinschaft der wissenschaftlichen medizinischen Fachgesellschaft (awmf) was carried out. Results: In addition to cycle irregularities that occur physiologically as a result of anovulatory cycles in the context of the immaturity of the hypothalamic–pituitary–gonadal axis, there are other cycle abnormalities that can be classified as pathological and need to be recognized and treated. Conclusions: Increasing awareness of the various specialist disciplines of physiological and pathological cycle abnormalities in adolescence and interdisciplinary cooperation between them can have a positive influence on the quality of life of adolescent women with cycle abnormalities. Full article
(This article belongs to the Section Obstetrics & Gynecology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>PRISMA flow diagram.</p>
Full article ">Figure 2
<p>Hormonal control of the hypothalamic–pituitary–ovary axis. GABA: gamma–aminobutyric acid.</p>
Full article ">Figure 3
<p>Abdominal sonography of an right (re) ovary in an adolescent girl (photo by the authors).</p>
Full article ">Figure 4
<p>Abdominal sonography of the messurement of the tissue of the uterus in an adolescent girl (photo by the authors).</p>
Full article ">Figure 5
<p>Abdominal sonography: Messurement of Excessively thickened endometrium in a 12-year-old girl with juvenile hypermenorrhea (photo by the authors).</p>
Full article ">Figure 6
<p>A 16-year-old girl with pronounced hirsutism and at high risk for PCOS (photo by the authors).</p>
Full article ">Figure 7
<p>Differential diagnoses of hyperandrogenemia.</p>
Full article ">
20 pages, 1362 KiB  
Systematic Review
Influence of Oestradiol Fluctuations in the Menstrual Cycle on Respiratory Exchange Ratio at Different Exercise Intensities: A Systematic Review, Meta-Analysis and Pooled-Data Analysis
by Catherine A. Rattley, Paul Ansdell, Louise C. Burgess, Malika Felton, Susan Dewhurst and Rebecca A. Neal
Physiologia 2024, 4(4), 486-505; https://doi.org/10.3390/physiologia4040033 - 16 Dec 2024
Viewed by 286
Abstract
Background: Oestradiol has been implicated as a factor in substrate utilisation in male and mouse studies but the effect of acute changes during the menstrual cycle is yet to be fully understood. Objective: To determine the role of oestradiol in respiratory exchange ratio [...] Read more.
Background: Oestradiol has been implicated as a factor in substrate utilisation in male and mouse studies but the effect of acute changes during the menstrual cycle is yet to be fully understood. Objective: To determine the role of oestradiol in respiratory exchange ratio (RER) during exercise at various intensities. Methods: This systematic review was conducted and reported in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. From inception to November 2023, four online databases (Cochrane, SPORTDiscus, MEDline and Web of Science) were searched for relevant articles. Studies that reported a resting oestradiol measurement in naturally menstruating women with exercise at a percentage of maximal aerobic capacity (%V˙O2max) were included. Mean and standard deviation for oestradiol, RER and exercise intensity were extracted and study quality assessed using a modified Downs and Black checklist. Risk of bias was assessed using I2 measure of heterogeneity and Egger’s regression test, assessment of bias from methodological quality was identified by sensitivity analysis. Eligible datasets were extracted for pairwise comparisons within a meta-analysis and correlation between change in oestradiol and change in RER. Data were also pooled to produce a mean and standard deviation for RER for menstrual stage and for low and high oestradiol groups. Results: Twenty-four articles were identified, over 50% were identified as high quality. Sixteen articles included datasets eligible for meta-analysis. Eleven articles utilised a submaximal constant-load exercise intensity, finding a standardised mean difference of − 0.09 ([CI: −0.35–0.17], p = 0.5) suggesting no effect of menstrual phase on constant-load exercise RER. In six articles using incremental exercise tests to exhaustion, a standardised mean difference of 0.60 ([CI 0.00–1.19], p = 0.05) was identified towards a higher maximal RER attained in follicular compared to luteal phase. There was no correlation (R = −0.26, p = 0.2) between change in oestradiol and change in RER between phases. All 24 articles, totalling 650 participants, were included in pooled analysis. When grouped by menstrual cycle phase or when grouped by oestradiol levels, RER was higher in the follicular phase than the luteal phase at low and high constant load exercise intensities. Discussion: Findings from the pooled-analysis and meta-analysis suggest that there may be menstrual cycle phase differences in RER that are intensity dependent. These differences may be related to sex hormone levels, but this was not supported by evidence of correlation between differences in RER and differences in oestradiol. At present, it remains best practice to assess performance in the same menstrual cycle phase if seeking to assess change from baseline. Full article
(This article belongs to the Section Exercise Physiology)
Show Figures

Figure 1

Figure 1
<p>Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) flow diagram for literature search, screening and selection.</p>
Full article ">Figure 2
<p>Quality of studies included in review and analysis (<span class="html-italic">n</span> = 24).</p>
Full article ">Figure 3
<p>(<b>a</b>) Forest plot of meta-analysis comparison of respiratory exchange ratio (RER) across all exercise intensities between early-mid follicular and early to mid-luteal phases up to 99%V˙O<sub>2max</sub>. Squares indicate the weight of standardized mean difference and 95% confidence intervals [CI]. Negative effect sizes indicate a higher RER observed in the luteal phase, and positive effect sizes indicate a higher RER observed in the follicular phase. * denotes studies removed for sensitivity analysis. Brackets indicate exercise intensity as a %V˙O<sub>2max</sub>. (<b>b</b>) Funnel plot of studies comparing respiratory exchange ratio between menstrual cycle phases with standardised mean difference plotted against standard error (13 paired datasets) [<a href="#B29-physiologia-04-00033" class="html-bibr">29</a>,<a href="#B30-physiologia-04-00033" class="html-bibr">30</a>,<a href="#B32-physiologia-04-00033" class="html-bibr">32</a>,<a href="#B34-physiologia-04-00033" class="html-bibr">34</a>,<a href="#B36-physiologia-04-00033" class="html-bibr">36</a>,<a href="#B37-physiologia-04-00033" class="html-bibr">37</a>,<a href="#B38-physiologia-04-00033" class="html-bibr">38</a>,<a href="#B45-physiologia-04-00033" class="html-bibr">45</a>,<a href="#B46-physiologia-04-00033" class="html-bibr">46</a>,<a href="#B50-physiologia-04-00033" class="html-bibr">50</a>,<a href="#B51-physiologia-04-00033" class="html-bibr">51</a>].</p>
Full article ">Figure 4
<p><b>Forest plot of meta-analysis comparison of respiratory exchange ratio</b> (RER) between early-mid follicular and early to mid-luteal phases at 100%V˙O<sub>2max</sub>. Squares indicate the weight of standardized mean difference and 95% confidence intervals [CI]. Negative effect sizes indicate a higher RER observed in the luteal phase, and positive effect sizes indicate higher RER observed in the follicular phase [<a href="#B33-physiologia-04-00033" class="html-bibr">33</a>,<a href="#B34-physiologia-04-00033" class="html-bibr">34</a>,<a href="#B35-physiologia-04-00033" class="html-bibr">35</a>,<a href="#B43-physiologia-04-00033" class="html-bibr">43</a>,<a href="#B49-physiologia-04-00033" class="html-bibr">49</a>,<a href="#B52-physiologia-04-00033" class="html-bibr">52</a>].</p>
Full article ">Figure 5
<p>Correlation between percent change in oestradiol and percent change in respiratory exchange ratio (<span class="html-italic">n</span> = 20 studies).</p>
Full article ">
6 pages, 1660 KiB  
Case Report
Difficult Diagnosis of Spontaneous Intracranial Hypotension with Nausea and Lower Abdominal Pain as Main Complaints: A Case Report
by Misaki Yokoi, Tsuneaki Kenzaka, Mari Asano, Ryu Sugimoto and Hogara Nishisaki
Reports 2024, 7(4), 115; https://doi.org/10.3390/reports7040115 - 16 Dec 2024
Viewed by 284
Abstract
Background and Clinical Significance: Symptoms of spontaneous intracranial hypotension include orthostatic headaches due to decreased cerebrospinal fluid (CSF) levels. Here, we present a 24-year-old female admitted to an obstetrics and gynecology department with primary complaints of lower abdominal pain and dysmenorrhea with [...] Read more.
Background and Clinical Significance: Symptoms of spontaneous intracranial hypotension include orthostatic headaches due to decreased cerebrospinal fluid (CSF) levels. Here, we present a 24-year-old female admitted to an obstetrics and gynecology department with primary complaints of lower abdominal pain and dysmenorrhea with subsequent diagnosis of spontaneous intracranial hypotension (SIH). Case Presentation: The patient had experienced nausea and lower abdominal pain independent of her menstrual cycle 5 days before admission, for which she visited the emergency department 3 days later. On admission, her symptoms were temporarily relieved by administering analgesics; thus, she was discharged. However, later, the symptoms worsened. Consequently, she returned to the emergency department for further evaluation, including blood tests, imaging, and endoscopy, which revealed no nausea- or abdominal pain-related organic abnormalities. On day 10, she developed a headache, aggravated by lying in the supine position and improved by sitting. Additional history revealed a diagnosis of SIH owing to the worsening abdominal pain in the supine position. An 111In CSF cavity scintigram showed no spinal fluid leakage; early intrabladder radioisotope (RI) accumulation was observed, and the residual 24 h CSF cavity RI was >30%. At a referral specialist hospital, an epidural saline infusion test was performed, which improved her headache and lower abdominal pain. Blood patch therapy improved her lower abdominal pain, headache, and dysmenorrhea. Conclusions: The final diagnosis was SIH, with symptoms attributed to CSF depletion. The patient also experienced rare paradoxical postural-related headaches and lower abdominal pain, aggravated by lying in the supine position, contributing to the final diagnosis. Full article
Show Figures

Figure 1

Figure 1
<p>Magnetic resonance imaging (MRI) of the head. (<b>a</b>) Non-contrast fluid-attenuated inversion recovery image showing no subdural fluid accumulation. (<b>b</b>) Contrast-enhanced T1-weighted image showing no diffuse dural thickening. (<b>c</b>) Contrast-enhanced T1-weighted fat-suppressed image showing no dilation of the epidural venous plexus. FLAIR, fluid-attenuated inversion recovery.</p>
Full article ">Figure 2
<p><sup>111</sup>In cerebrospinal fluid cavity scintigrams at 1 h. No spinal fluid leakage was observed. However, early accumulation of radioisotopes in the bladder was observed, suggesting spinal fluid leakage.</p>
Full article ">Figure 3
<p><sup>111</sup>In cerebrospinal fluid cavity scintigrams at 24 h. The residual radioisotope in the cerebrospinal fluid space was 15% at 24 h (normal, &gt;30%), suggesting spinal fluid leakage.</p>
Full article ">Figure 4
<p>Schematic diagram of spinal nerve root traction associated with SIH and nerve root compression by a dilated epidural venous plexus. Original drawing based on ref. [<a href="#B6-reports-07-00115" class="html-bibr">6</a>].</p>
Full article ">
8 pages, 1318 KiB  
Article
Prolactin Response to a Submaximal Dose of Ghrelin in Different Phases of the Normal Menstrual Cycle
by Christina I. Messini, George Anifandis, Panagiotis Georgoulias, Konstantinos Dafopoulos, George Sveronis, Alexandros Daponte and Ioannis E. Messinis
Medicina 2024, 60(12), 2039; https://doi.org/10.3390/medicina60122039 - 11 Dec 2024
Viewed by 378
Abstract
Background and Objectives: A similar secretory pattern of prolactin (PRL) and growth hormone (GH) during the menstrual cycle has been reported in response to a high dose of ghrelin in adult healthy women. The present study aimed to assess the pattern of [...] Read more.
Background and Objectives: A similar secretory pattern of prolactin (PRL) and growth hormone (GH) during the menstrual cycle has been reported in response to a high dose of ghrelin in adult healthy women. The present study aimed to assess the pattern of PRL and GH secretions in response to a submaximal dose of ghrelin during different menstrual phases in adult healthy women. Materials and Methods: Eight female subjects with normal cyclicity were enrolled. These subjects were either in the early follicular (EF), late follicular (LF), or mid-luteal (ML) phase of their cycles. Each subject received an IV dose of normal saline (2 mL each time) during the first cycle after enrollment, followed by an IV dose of ghrelin (0.30 μg/kg bw) in the second cycle. The blood samples were collected before and after the IV dosage at −15, 0, 15, 30, 45, 60, 75, 90 and 120 min, where 0 min denotes the time of IV dosage. Results: All the enrolled subjects experienced ovulatory cycles as assessed by increased serum progesterone levels. Serum estradiol levels were significantly higher in the LF than in the EF (p < 0.001) and ML phases (p < 0.01); these levels were also significantly higher in the ML than in the EF phase (p < 0.01). The administration of saline did not affect serum GH or PRL levels. Following the administration of ghrelin, plasma ghrelin levels and serum GH levels increased significantly (p < 0.001). The response amplitude of GH was similar in the three stages of cycle 2. In contrast to GH, the ghrelin injection induced a significant increase in serum PRL levels only in the LF phase (p < 0.05). Conclusions: These results show, for the first time, a different pattern of PRL and GH in response to a submaximal dose of ghrelin during the normal menstrual cycle. It is suggested that the ghrelin threshold for pituitary lactotrophs is higher than for somatotrophs and that, unlike GH, ghrelin-stimulated PRL secretion can be influenced by ovarian steroids. Full article
(This article belongs to the Section Endocrinology)
Show Figures

Figure 1

Figure 1
<p>Plasma ghrelin values (pg/mL) before and after an acute IV injection (arrow) of normal saline (2 mL) to 8 healthy women during the (◊) early follicular phase (EF) of cycle 1 (control) as well as before and after an acute IV injection (arrow) of ghrelin (0.30 μg/kg) to the same women during the (○) EF, (●) late follicular (LF) and (♦) mid-luteal phase (ML) of cycle 2. ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 (difference from cycle 1).</p>
Full article ">Figure 2
<p>Serum growth hormone (GH) values (ng/mL) before and after an acute IV injection (arrow) of normal saline (2 mL) to 8 healthy women during the (◊) early follicular phase (EF) of cycle 1 (control) as well as before and after an acute IV injection (arrow) of ghrelin (0.30 μg/kg) to the same women during the (○) EF, (●) late follicular (LF) and (♦) mid-luteal phase (ML) of cycle 2. ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 (difference from cycle 1).</p>
Full article ">Figure 3
<p>Serum prolactin (PRL) values (ng/mL) before and after an acute IV injection (arrow) of normal saline (2 mL) to 8 healthy women during the (◊) early follicular phase (EF) of cycle 1 (control) as well as before and after an acute IV injection (arrow) of ghrelin (0.30 μg/kg) to the same women during the (○) (EF), (●) late follicular (LF) and (♦) mid-luteal phase (MF) of cycle 2. * <span class="html-italic">p</span> &lt; 0.05 (difference from cycle 1).</p>
Full article ">
14 pages, 318 KiB  
Review
The Interplay of Uterine Health and Obesity: A Comprehensive Review
by Dina Šišljagić, Senka Blažetić, Marija Heffer, Mihaela Vranješ Delać and Andrijana Muller
Biomedicines 2024, 12(12), 2801; https://doi.org/10.3390/biomedicines12122801 - 10 Dec 2024
Viewed by 518
Abstract
Uterine physiology encompasses the intricate processes governing the structure, function, and regulation of the uterus, a pivotal organ within the female reproductive system. The escalating prevalence of obesity has emerged as a significant global health issue, profoundly impacting various facets of well-being, including [...] Read more.
Uterine physiology encompasses the intricate processes governing the structure, function, and regulation of the uterus, a pivotal organ within the female reproductive system. The escalating prevalence of obesity has emerged as a significant global health issue, profoundly impacting various facets of well-being, including female reproductive health. These effects extend to uterine structure and function, influencing reproductive health outcomes in women. They encompass alterations in uterine morphology, disruptions in hormonal signaling, and inflammatory processes. Insulin and leptin, pivotal hormones regulating metabolism, energy balance, and reproductive function, play crucial roles in this context. Insulin chiefly governs glucose metabolism and storage, while leptin regulates appetite and energy expenditure. However, in obesity, resistance to both insulin and leptin can develop, impacting uterine function. Inflammation and oxidative stress further exacerbate the development of uterine dysfunction in obesity. Chronic low-grade inflammation and heightened oxidative stress, characteristic of obesity, contribute to metabolic disruptions and tissue damage, including within the uterus. Obesity significantly disrupts menstrual cycles, fertility, and pregnancy outcomes in women. The accumulation of excess adipose tissue disrupts hormonal equilibrium, disturbs ovarian function, and fosters metabolic irregularities, all of which detrimentally impact reproductive health. Full article
(This article belongs to the Special Issue Molecular Research in Obesity)
17 pages, 279 KiB  
Article
Just 4U™: Reusable Period Pants Alongside an Ovulatory Menstrual Health Literacy Program
by Kate Fraser Roux, Felicity Roux, Jacqueline Hendriks, HuiJun Chih and Sharyn Burns
Youth 2024, 4(4), 1757-1773; https://doi.org/10.3390/youth4040111 - 9 Dec 2024
Viewed by 416
Abstract
The concept of “period poverty” describes the lack of access to menstrual health education and menstrual care products. This quasi-experimental mixed-methods study evaluated a collaboration called Just 4U™ to address period poverty. This collaboration was formed between My Vital Cycles®, [...] Read more.
The concept of “period poverty” describes the lack of access to menstrual health education and menstrual care products. This quasi-experimental mixed-methods study evaluated a collaboration called Just 4U™ to address period poverty. This collaboration was formed between My Vital Cycles®, as the provider of educational content, and Modibodi®, as the provider of period pants as a reusable menstrual product (RMP). Five co-educational schools, including a regional school, participated and were of average to below-average socio-educational advantage ranking in Australia. The pre- and post-intervention evaluation with 63 postmenarcheal adolescents (14–18-year-old) in Grades 9–12 showed an improvement in their ovulatory menstrual health literacy. Open-ended questions explored their perspectives on the RMP. Overall, the RMP was well received by participants, who reported a positive impact on their mindset and cycle management. School staff (n = 6) who had observed delivery of Just 4U™ were interviewed. They believed the program had benefited the participants and recommended that teacher training would help the intervention to be sustainably implemented in schools. Findings highlight the worthwhile inclusion of RMPs alongside ovulatory menstrual health education. This study contributes to ongoing research in adolescent ovulatory menstrual health education. Full article
(This article belongs to the Special Issue Sexuality: Health, Education and Rights)
9 pages, 1826 KiB  
Review
Catamenial Pneumothorax—Still an Unveiled Disease
by Iwona Damps-Konstańska, Adriana Szukalska, Piotr Janowiak and Ewa Jassem
Medicina 2024, 60(12), 2029; https://doi.org/10.3390/medicina60122029 - 9 Dec 2024
Viewed by 416
Abstract
This review presents current opinions on an uncommon condition called catamenial pneumothorax (CP), which is usually associated with thoracic endometriosis syndrome (TES). TES is characterized by the presence of endometriotic lesions in pleura and lung parenchyma and presents with various clinical signs and [...] Read more.
This review presents current opinions on an uncommon condition called catamenial pneumothorax (CP), which is usually associated with thoracic endometriosis syndrome (TES). TES is characterized by the presence of endometriotic lesions in pleura and lung parenchyma and presents with various clinical signs and symptoms, including catamenial pneumothorax. Their diagnosis is often delayed. Pulmonary endometric lesions, however, often detected in patients with hemothorax and hemoptysis, may be absent in a proportion of cases of pneumothorax. The typical presentation of CP includes signs and symptoms of pneumothorax, which occur along with menstruation, most commonly around 24 h before and 48–72 h after its onset. However, they may not occur during every menstrual cycle. Suggestive CP lesions on conventional radiography (RTG) include pneumoperitoneum accompanying right-sided pneumothorax, lung opacities, pleural effusion, and nodular infiltrates. Chest and abdomen computed tomography (CT), particularly contrast-enhanced, may additionally show pneumoperitoneum and diaphragmatic lesions. The management of CP includes supportive treatment of acute symptoms and causal treatment to prevent recurrent disease. This article presents the pathophysiology of CP, an overview of the diagnostic methods, and the current therapeutic approaches. The necessity for a multidisciplinary approach to the diagnosis of CP and to the choice of the best treatment modality is underlined (promising new therapeutic options are also mentioned); however, international guidelines are still missing. Full article
(This article belongs to the Section Pulmonology)
Show Figures

Figure 1

Figure 1
<p>Diagnostic schema.</p>
Full article ">Figure 2
<p>Treatment strategy.</p>
Full article ">Figure 3
<p>Diagnostic and therapeutic pathway.</p>
Full article ">
12 pages, 863 KiB  
Article
A Ketogenic Diet Followed by Gradual Carbohydrate Reintroduction Restores Menstrual Cycles in Women with Polycystic Ovary Syndrome with Oligomenorrhea Independent of Body Weight Loss: Results from a Single-Center, One-Arm, Pilot Study
by Rebecca Rossetti, Vittoria Strinati, Alessandra Caputi, Renata Risi, Maria Letizia Spizzichini, Alessandro Mondo, Lorenzo Spiniello, Carla Lubrano, Antonella Giancotti, Dario Tuccinardi, Lucio Gnessi and Mikiko Watanabe
Metabolites 2024, 14(12), 691; https://doi.org/10.3390/metabo14120691 - 8 Dec 2024
Viewed by 1255
Abstract
Background/Objectives: Polycystic ovary syndrome (PCOS) is a common endocrine disorder in women of fertile age. Some studies suggest that a ketogenic diet (KD) may have a role in treating PCOS. We aimed to demonstrate the long-term effectiveness of a KD in PCOS. [...] Read more.
Background/Objectives: Polycystic ovary syndrome (PCOS) is a common endocrine disorder in women of fertile age. Some studies suggest that a ketogenic diet (KD) may have a role in treating PCOS. We aimed to demonstrate the long-term effectiveness of a KD in PCOS. Methods: Eighteen patients with PCOS phenotype A were enrolled: 28% were of normal weight, 28% were overweight, and 44% had obesity. All participants followed a KD without meal replacements for 45 days. After this period, patients underwent gradual carbohydrate reintroduction over 45 days, and thereafter healthy eating indications were given. Twelve patients completed the study. The patients were assessed at baseline and after 6 months. Anthropometric data, body composition, pelvic ultrasound, blood chemistry, hirsutism, and menstrual cycles frequency were recorded; Results: Besides improvement in anthropometric parameters, menstrual cycles (p 0.012), ovarian volume (p 0.029), FSH (p 0.05), LH (p 0.037), and progesterone (p 0.017) improved independently of weight or fat loss. However, testosterone and hirsutism improvements were influenced by weight and fat mass reduction. Conclusions: Our study showed that a KD followed by gradual carbohydrate reintroduction in PCOS has beneficial effects medium term, mostly independent of body weight loss, even in normal-weight women, suggesting that nutritional ketosis exerts beneficial effects per se. Full article
Show Figures

Figure 1

Figure 1
<p>Changes in (<b>A</b>) body weight, (<b>B</b>) waist circumference, (<b>C</b>) hip circumference, (<b>D</b>) capillary beta-hydroxybutyrate (BHB) over time. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>Menstrual cycle frequency of each patient who completed the study in the six months prior to study start and during the study up to the end of follow up. Each horizontal line represents one patient, each vertical line represents one bleeding. KD, ketogenic diet; LCD, low carbohydrate diet.</p>
Full article ">
19 pages, 7854 KiB  
Article
Single-Cell RNA Sequencing of PBMCs Identified Junction Plakoglobin (JUP) as Stratification Biomarker for Endometriosis
by Thomas Andrieu, Angelo Duo, Lea Duempelmann, Magdalena Patzak, Flurina Annacarina Maria Saner, Jitka Skrabalova, Cinzia Donato, Peter Nestorov and Michael D. Mueller
Int. J. Mol. Sci. 2024, 25(23), 13071; https://doi.org/10.3390/ijms252313071 - 5 Dec 2024
Viewed by 532
Abstract
This study aimed to identify unique characteristics in the peripheral blood mononuclear cells (PBMCs) of endometriosis patients and develop a non-invasive early diagnostic tool. Using single-cell RNA sequencing (scRNA-seq), we constructed the first single-cell atlas of PBMCs from endometriosis patients based on 107,964 [...] Read more.
This study aimed to identify unique characteristics in the peripheral blood mononuclear cells (PBMCs) of endometriosis patients and develop a non-invasive early diagnostic tool. Using single-cell RNA sequencing (scRNA-seq), we constructed the first single-cell atlas of PBMCs from endometriosis patients based on 107,964 cells and 25,847 genes. Within CD16+ monocytes, we discovered JUP as a dysregulated gene. To assess its diagnostic potential, we measured peritoneal fluid (PF) and serum JUP levels in a large cohort of 199 patients including 20 women with ovarian cancer (OC). JUP was barely detectable in PF but was significantly elevated in the serum of patients with endometriosis and OC, with levels 1.33 and 2.34 times higher than controls, respectively. Additionally, JUP was found in conditioned culture media of CD14+/CD16+ monocytes aligning with our scRNA-seq data. Serum JUP levels correlated with endometriosis severity and endometrioma presence but were unaffected by dysmenorrhea, menstrual cycle, or adenomyosis. When combined with CA125 (cancer antigen 125) JUP enhanced the specificity of endometriosis diagnosis from 89.13% (CA125 measured alone) to 100%. While sensitivity remains a challenge at 19%, our results suggest that JUP’s potential to enhance diagnostic accuracy warrants additional investigation. Furthermore, employing serum JUP as a stratification marker unlocked the potential to identify additional endometriosis-related genes, offering novel insights into disease pathogenesis. Full article
(This article belongs to the Special Issue Biomarkers and Early Detection Strategies of Ovarian Tumors)
Show Figures

Figure 1

Figure 1
<p>Dysregulated gene expression analysis with endometriosis as endpoint. (<b>A</b>) Study design. Pain scores and peripheral blood were collected just before laparoscopy. PBMCs were cryopreserved until single-cell sequencing could be performed by batch. Serums were frozen before quantification of JUP, CA125 and S100A12 by ELISA. During surgery, the revised American Society for Reproductive Medicine (rASRM) score was assigned and suspect lesions were collected for confirmation of diagnosis by a trained pathologist. DGE (differential gene expression) analysis was based on endometriosis diagnosis and level of JUP in serum. (Created in <a href="https://BioRender.com" target="_blank">https://BioRender.com</a>, accessed on 8 November 2024) (<b>B</b>) Cellular composition of PBMCs based on scRNA-seq (uniform manifold approximation and projection, UMAP plot) for all samples (controls = 7; endometriosis cases = 6). (<b>C</b>) Dot plot showing average expression (color scale = expression intensity) and percentage of positive cells (dimension scale = proportion of positive cells) for selected cell type-specific marker genes. (<b>D</b>) Comparison of JUP expression profile in endometriosis cases and controls. The left panel represents the expression level of JUP in CD16<sup>+</sup> monocytes (logFC 1.66; adjusted <span class="html-italic">p</span>-value = 0.004). The right panel represents the UMAP of the CD16<sup>+</sup> monocytes in samples of women without endometriosis (left) and with endometriosis (right). Cells with higher expression levels are indicated by darker dots. Endo: Endometriosis group, CD4 TCM: CD4 central memory, CD4 TEM: CD4 effector memory T cells, CD8 TCM: CD8 central memory, CD8 TEM: CD8 effector memory T cells, cDC2: Type-2 conventional dendritic cells, dnT: TCRα<sup>+</sup> CD4<sup>−</sup> CD8<sup>−</sup> double negative T cells, Eryth: erythrocytes, gdT (Yδt): Gamma-delta (γδ) T cells, HSPC: Hematopoietic Stem and Progenitor Cells, MAIT: Mucosal-Associated Invariant T cells, NK: Natural killer cells, pDC: Plasmacytoid dendritic cells, Treg: regulatory T cells.</p>
Full article ">Figure 2
<p>JUP in endometriosis. (<b>A</b>) Violin plots of serum JUP level in endometriosis-free women (CTL) (median = 158.5 ng/mL), endometriosis and patients (Endo) (median = 211.4 ng/mL), patients with ovarian cancer (OC) (median = 372.3 ng/mL). CTL vs. Endo (<span class="html-italic">p</span>-value = 0.0152), CTL vs. OC (<span class="html-italic">p</span>-value &lt; 0.0001), Endo vs. OC (<span class="html-italic">p</span>-value = 0.0027) (Mann–Whitney test). (<b>B</b>) Violin plots of serum JUP level in CTL (median = 158.5 ng/mL), Endo stage I–II (median = 199.5 ng/mL) and Endo stage III-IV (median = 283.4 ng/mL). Non-parametric one-way ANOVA (analysis of variance, Kruskal–Wallis test, <span class="html-italic">p</span>-value = 0.0184) followed by a non-parametric comparison (Dunn’s multiple comparisons test, CTL vs. Endo stage III-IV, <span class="html-italic">p</span>-value = 0.019). Patients with adenomyosis were included. (<b>C</b>) Violin plots of serum JUP level in CTL (median = 158.5 ng/mL), endometriosis patients without endometrioma (Endo WO OMA) (median = 203.1 ng/mL), and endometriosis patients with endometrioma (Endo with OMA) (median = 283.4 ng/mL). Non-parametric one-way ANOVA (Kruskal-Wallis test, <span class="html-italic">p</span>-value = 0.007) followed by a non-parametric comparison (Dunn’s multiple comparisons test, CTL vs. Endo stage III-IV, <span class="html-italic">p</span>-value = 0.006). Patients with adenomyosis were included. (<b>D</b>) Violin plots of serum JUP level in endometriosis-free women (CTL) (median = 165.5 ng/mL), endometriosis patients (Endo) (median = 207.4 ng/mL), endometriosis-free women with adenomyosis (Adeno) (median = 139.5 ng/mL), and endometriosis patients with adenomyosis (Endo and Adeno) (median = 255.9 ng/mL). Two-way ANOVA assessing the endometriosis and adenomyosis effects (endometriosis, <span class="html-italic">p</span>-value = 0.0151; adenomyosis, <span class="html-italic">p</span>-value = 0.1167). (<b>E</b>) Violin plots of serum JUP level in proliferative phase (CTL Prolif: median = 166.5 ng/mL; Endo Prolif: median = 116.8 ng/mL) and in secretory phase (CTL Secr: median = 162.2 ng/mL and Endo Secr: median = 215.5 ng/mL). Two-way ANOVA assessing the endometriosis and cycle effects (endometriosis, <span class="html-italic">p</span>-value = 0.0345; cycle, <span class="html-italic">p</span>-value = 0.2251). (<b>F</b>) Positive correlation of serum JUP level with BMI (Body Mass Index; Pearson’s r = 0.260, <span class="html-italic">p</span>-value = 0.003, and n = 128). (* = <span class="html-italic">p</span>-value &lt; 0.05, ** = <span class="html-italic">p</span>-value &lt; 0.01, *** = <span class="html-italic">p</span>-value &lt; 0.001, n.s. = not significant).</p>
Full article ">Figure 3
<p>Diagnosis performance of JUP in endometriosis. (<b>A</b>) ROC analysis for serum JUP (vertical lines, AUC (Area Under the Curve) 0.6048, <span class="html-italic">p</span>-value = 0.0154) and serum CA125 (clear circles, AUC 0.7054, <span class="html-italic">p</span>-value = 0.0006). JUP AUC vs. CA125 AUC (<span class="html-italic">p</span>-value = 0.1476). (<b>B</b>) Positive correlation of JUP with CA125 in serum; Pearson’s r = 0.551; <span class="html-italic">p</span>-value 1.020 × 10<sup>−8</sup>; n = 93 (CTL: clear circles; endometriosis: black circles). (<b>C</b>) Box and Whiskers plots of HE4 (Human Epididymis Protein 4) quantified in serum in patients with high CA125 (&gt;26.8 U/mL) and high JUP (&gt;324 ng/mL). HE4 was higher in patients with ovarian cancer (median = 189.0 pM) than in patients with endometriosis (median = 26.7 pM) (<span class="html-italic">p</span>-value &lt; 0.0001, Mann-Whitney test). (<b>D</b>) Positive correlation of JUP with S100A12 in serum; Pearson’s r = 0.835; <span class="html-italic">p</span>-value = 2.180 × 10<sup>−16</sup>; n = 59 (CTL: clear circles; endometriosis: black circles). (*** = <span class="html-italic">p</span>-value &lt; 0.001).</p>
Full article ">Figure 4
<p>JUP and S100A12 levels in conditioned medium of CD14<sup>+</sup>/CD16<sup>+</sup> monocytes in culture. (<b>A</b>) Box and Whiskers plots of S100A12 quantified in medium at 24 h (median = 6.50 ng/mL), 72 h (median = 2.90 ng/mL), and 120 h (median = 1.00 ng/mL) of in vitro culture. One-way ANOVA (Friedman paired test) (<span class="html-italic">p</span>-value = 0.0028). and 24 h vs. 120 h (<span class="html-italic">p</span>-value = 0.0073) (Dunn’s multiple comparisons test). (<b>B</b>) Box and Whiskers plots of JUP quantified in medium at 24 h (median = 1.83 ng/mL), 72 h (median = 1.70 ng/mL), and 120 h (median = 0.96 ng/mL) of in vitro culture. One-way ANOVA (Friedman paired test) (<span class="html-italic">p</span>-value = 0.0054), 24 h vs. 120 h (<span class="html-italic">p</span>-value = 0.0424), and 72 h vs. 120 h (<span class="html-italic">p</span>-value = 0.0183) (Dunn’s multiple comparisons test). (<b>C</b>) Positive correlation of JUP with S100A12 in culture medium; Pearson’s r = 0.889; <span class="html-italic">p</span> &lt; 0.0001; n = 18. The values collected at 24 h, 96 h and 120 h for the 6 PBMC cultures were included in this correlation plot. (* = <span class="html-italic">p</span>-value &lt; 0.05, ** = <span class="html-italic">p</span>-value &lt; 0.01).</p>
Full article ">Figure 5
<p>Expression profile of endometriosis DEGs in patients with high (<b>A</b>–<b>C</b>) and low (<b>D</b>–<b>F</b>) serum JUP. The upper panels represent the expression level of the DEGs in each identified cluster; (<b>A</b>) <span class="html-italic">RGPD2</span> in CD14<sup>+</sup> monocytes; (<b>B</b>) <span class="html-italic">LTB</span> in γδt; (<b>C</b>) <span class="html-italic">KLRC1</span> in γδt; (<b>D</b>) <span class="html-italic">TMEM176A</span> in CD14<sup>+</sup> monocytes; (<b>E</b>) <span class="html-italic">TMEM176B</span> in CD14<sup>+</sup> monocytes; and (<b>F</b>) <span class="html-italic">TRBV2</span> in CD4 CTLs. The lower panels represent the UMAP of each identified cluster in samples of women without endometriosis (left) and with endometriosis (right). A color code indicates the expression level of the DEGs in each cell; cells with higher expression levels are indicated by darker dots. <span class="html-italic">KLRC1</span>: Killer cell lectin-like receptor C1, <span class="html-italic">LTB</span>: lymphotoxin beta, <span class="html-italic">RGPD2</span>: RANBP2-like and GRIP domain-containing 2, <span class="html-italic">TMEM176A</span> and <span class="html-italic">B</span>: Transmembrane proteins 176A and B, <span class="html-italic">TRBV2</span>: T Cell Receptor Beta Variable 2.</p>
Full article ">
42 pages, 3923 KiB  
Review
Environmental Exposure to Per- and Polyfluorylalkyl Substances (PFASs) and Reproductive Outcomes in the General Population: A Systematic Review of Epidemiological Studies
by Alex Haimbaugh, Danielle N. Meyer, Mackenzie L. Connell, Jessica Blount-Pacheco, Dienye Tolofari, Gabrielle Gonzalez, Dayita Banerjee, John Norton, Carol J. Miller and Tracie R. Baker
Int. J. Environ. Res. Public Health 2024, 21(12), 1615; https://doi.org/10.3390/ijerph21121615 - 2 Dec 2024
Viewed by 1143
Abstract
This Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) systematic review synthesized effects of background levels of per- and polyfluorylalkyl substance (PFAS) levels on reproductive health outcomes in the general public: fertility, preterm birth, miscarriage, ovarian health, menstruation, menopause, sperm health, and [...] Read more.
This Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) systematic review synthesized effects of background levels of per- and polyfluorylalkyl substance (PFAS) levels on reproductive health outcomes in the general public: fertility, preterm birth, miscarriage, ovarian health, menstruation, menopause, sperm health, and in utero fetal growth. The inclusion criteria included original research (or primary) studies, human subjects, and investigation of outcomes of interest following non-occupational exposures. It drew from four databases (Web of Science, PubMed, Embase and Health and Environmental Research Online (HERO)) using a standardized search string for all studies published between 1 January 2017 and 13 April 2022. Risk of bias was assessed by two independent reviewers. Data were extracted and reviewed by multiple reviewers. Each study was summarized under its outcome in terms of methodology and results and placed in context, with recommendations for future research. Of 1712 records identified, 30 were eligible, with a total of 27,901 participants (33 datasets, as three studies included multiple outcomes). There was no effect of background levels of PFAS on fertility. There were weakly to moderately increased odds of preterm birth with higher perfluorooctane sulfonic acid (PFOS) levels; the same for miscarriage with perfluorooctanoic acid (PFOA) levels. There was limited yet suggestive evidence for a link between PFAS and early menopause and primary ovarian insufficiency; menstrual cycle characteristics were inconsistent. PFAS moderately increased odds of PCOS- and endometriosis-related infertility, respectively. Sperm motility and DNA health were moderately impaired by multiple PFAS. Fetal growth findings were inconsistent. This review may be used to inform forthcoming drinking water standards and policy initiatives regarding PFAS compounds and drinking water. Future reviews would benefit from more recent studies. Larger studies in these areas are warranted. Future studies should plan large cohorts and open access data availability to capture small effects and serve the public. Funding: Great Lakes Water Authority (Detroit, MI), the Erb Family Foundation through Healthy Urban Waters at Wayne State University (Detroit, MI), and Wayne State University CLEAR Superfund Research (NIH P42ES030991). Full article
(This article belongs to the Section Environmental Health)
Show Figures

Figure 1

Figure 1
<p>PRISMA flow diagram.</p>
Full article ">Figure 2
<p>Pie charts depicting study characteristics. Percentages are rounded to nearest whole number and may not add up to 100%. (<b>a</b>) Number of studies included for each outcome. Some studies measured multiple outcomes. (<b>b</b>) Media type used in studies. Some studies used multiple media. (<b>c</b>) Study design type. (<b>d</b>) Open access status. (<b>e</b>) Region of studies. Scandinavia includes Sweden, Norway, Faroe Islands, and Denmark.</p>
Full article ">Figure 3
<p>(<b>a</b>) Participants in each outcome. (<b>b</b>) Average risk of bias scores in each outcome stratified by cohort or cross-sectional studies (left) or case–control (right). Minimum points to be included in the review for cohort/cross-sectional was 7; maximum achievable was 10. Minimum points to be included in the review for case–control was 10; maximum achievable was 14. (<b>c</b>) Median PFAS levels (ng/mL) in each study in maternal/paternal blood, serum, or plasma. Levels reported from controls only in case–control studies. Multi-region studies denoted with (S) for Sweden, (N) for Norway, (Y) for Yantai, and (B) for Beijing. Meng et al., 2018 [<a href="#B58-ijerph-21-01615" class="html-bibr">58</a>]. Sagiv et al., 2017 [<a href="#B59-ijerph-21-01615" class="html-bibr">59</a>]. Liew et al., 2020 [<a href="#B60-ijerph-21-01615" class="html-bibr">60</a>]. Petersen et al., 2018 [<a href="#B61-ijerph-21-01615" class="html-bibr">61</a>]. Ding et al., 2020 [<a href="#B62-ijerph-21-01615" class="html-bibr">62</a>]. Lauritzen et al., 2017 [<a href="#B63-ijerph-21-01615" class="html-bibr">63</a>]. Kalloo et al., 2020 [<a href="#B64-ijerph-21-01615" class="html-bibr">64</a>]. Singer et al., 2018 [<a href="#B65-ijerph-21-01615" class="html-bibr">65</a>]. Zhou et al., 2017 [<a href="#B66-ijerph-21-01615" class="html-bibr">66</a>]. Song et al., 2018 [<a href="#B67-ijerph-21-01615" class="html-bibr">67</a>]. Huo et al., 2020 [<a href="#B68-ijerph-21-01615" class="html-bibr">68</a>]. Pan et al., 2019 [<a href="#B69-ijerph-21-01615" class="html-bibr">69</a>]. Chu et al., 2020 [<a href="#B70-ijerph-21-01615" class="html-bibr">70</a>]. Wang et al., 2021 [<a href="#B71-ijerph-21-01615" class="html-bibr">71</a>]. Wang et al., 2017 [<a href="#B72-ijerph-21-01615" class="html-bibr">72</a>]. Costa et al., 2019 [<a href="#B73-ijerph-21-01615" class="html-bibr">73</a>]. Manzano-Salgado et al., 2017 [<a href="#B74-ijerph-21-01615" class="html-bibr">74</a>]. Zhang et al., 2018 [<a href="#B75-ijerph-21-01615" class="html-bibr">75</a>]. Wikström et al., 2021 [<a href="#B76-ijerph-21-01615" class="html-bibr">76</a>]. Ouidir et al., 2020 [<a href="#B77-ijerph-21-01615" class="html-bibr">77</a>]. Wang et al., 2021 [<a href="#B71-ijerph-21-01615" class="html-bibr">71</a>]. Wise et al., 2022 [<a href="#B78-ijerph-21-01615" class="html-bibr">78</a>]. Wang et al., 2019 [<a href="#B79-ijerph-21-01615" class="html-bibr">79</a>]. Bjorvang et al., 2022 [<a href="#B80-ijerph-21-01615" class="html-bibr">80</a>]. Heffernan et al., 2018 [<a href="#B81-ijerph-21-01615" class="html-bibr">81</a>]. Bjorvang et al., 2021 [<a href="#B82-ijerph-21-01615" class="html-bibr">82</a>]. Eick &amp; Hom Thepaksorn et al., 2020 [<a href="#B83-ijerph-21-01615" class="html-bibr">83</a>]. Liu et al., 2020 [<a href="#B84-ijerph-21-01615" class="html-bibr">84</a>]. Asterisk denotes the concentration of controls in case-control studies, if overall median is not reported.</p>
Full article ">Figure 4
<p>Forest plot of odds and risk ratios for preterm birth with increasing PFAS levels from four studies. LNE: line of no effect. PFNA: perfluorononanoic acid. PFHxS: perfluorohexane sulfonic acid. PFOS: perfluorooctane sulfonic acid. PFOA: perflurooctanoic acid. Yang et al., 2022 [<a href="#B108-ijerph-21-01615" class="html-bibr">108</a>]. Sagiv et al., 2018 [<a href="#B59-ijerph-21-01615" class="html-bibr">59</a>]. Manzano-Salgado et al., 2017 [<a href="#B74-ijerph-21-01615" class="html-bibr">74</a>]. Eick &amp; Hom Thepaksorn et al., 2020 [<a href="#B83-ijerph-21-01615" class="html-bibr">83</a>]. Liu et al., 2020 [<a href="#B84-ijerph-21-01615" class="html-bibr">84</a>]. Chu et al., 2020 [<a href="#B70-ijerph-21-01615" class="html-bibr">70</a>].</p>
Full article ">Figure 5
<p>Forest plot of miscarriage odds and risk ratios with increasing PFAS levels from three studies. LNE: line of no effect. PFOS: perfluorooctane sulfonic acid. PFOA: perflurooctanoic acid. PFNA: perfluorononanoic acid. PFHxS: perfluorohexane sulfonic acid. PFDA: perfluorodecanoic acid. Wang et al. (2021) results are from Beijing and Yantai sites combined. Wang et al., 2021 [<a href="#B71-ijerph-21-01615" class="html-bibr">71</a>]. Wikström et al., 2021 [<a href="#B76-ijerph-21-01615" class="html-bibr">76</a>]. Liew et al., 2020 [<a href="#B60-ijerph-21-01615" class="html-bibr">60</a>].</p>
Full article ">Figure 6
<p>Forest plot of odds and risk ratios of ovarian health effects (top: endometriosis; bottom: PCOS-related infertility) with increasing PFAS levels from two studies. PFHpA: perfluoroheptanoic acid. PFBS: perfluorobutanesulfonic acid. PFDoA: perfluorododecanoic acid. PFHxS: perfluorohexane sulfonic acid. PFUA: perfluoroundecanoic acid. PFDA: perfluorodecanoic acid. PFNA: perfluorononanoic acid. PFOS: perfluorooctanesulfonic acid. PFOA: perfluorooctanoic acid. Wang et al., 2019 [<a href="#B184-ijerph-21-01615" class="html-bibr">184</a>]. Wang et al., 2017 [<a href="#B72-ijerph-21-01615" class="html-bibr">72</a>].</p>
Full article ">Figure 7
<p>Forest plot of beta coefficients for sperm health effects with increasing PFAS levels in semen from two studies. Top panel: DNA stability (decreasing); DNA fragmentation index. Bottom panel: sperm motility (% motile sperm). PFDA: perfluorodecanoic acid. PFUnDA: perfluoroundecanoic acid. 6:2 Cl-PFESA: 6:2 chlorinated polyfluorinated ether sulfonate. PFNA: perfluorononanoic acid. PFOS: perfluorooctanesulfonic acid. PFOA: perfluorooctanoic acid. Pan et al., 2019 [<a href="#B69-ijerph-21-01615" class="html-bibr">69</a>]. Petersen et al., 2018 [<a href="#B61-ijerph-21-01615" class="html-bibr">61</a>].</p>
Full article ">Figure 8
<p>For each PFAS, median level (ng/mL) across ethnicities (concentrations are compared by column, not row). Color darkens with increasing median concentration. Me-FOSAA: N-methylperfluorooctane sulfonamidoacetic acid. PFDA: perfluorodecanoic acid. PFDoDA: perfluorododecanoic acid. PFHxS: perfluorohexane sulfonate. PFNA: perfluorononanoic acid. PFOA: perfluorooctanoic acid. PFOS: perfluorooctane sulfonate. PFUnDA: perfluoroundecanoic acid.</p>
Full article ">Figure 9
<p>Heatmap of beta-values at FDR &lt; 0.05 for measures of fetal growth, broken down by group. Warmer colors (orange, red) indicate lower values, yellow indicates mid-range values, and green indicates higher values. AC: abdominal circumference, FL: femur length, EFW: estimated fetal weight, BD: biparietal diameter. Head circumference not included (not significant for any ethnicity). Me-FOSAA: N-methylperfluorooctane sulfonamidoacetic acid. PFDA: perfluorodecanoic acid. PFDoDA: perfluorododecanoic acid. PFHxS: perfluorohexane sulfonate. PFNA: perfluorononanoic acid. PFOA: perfluorooctanoic acid. PFOS: perfluorooctane sulfonate. PFUnDA: perfluoroundecanoic acid.</p>
Full article ">
13 pages, 2313 KiB  
Article
Lactobacillus helveticus HY7801 Improves Premenstrual Syndrome Symptoms by Regulating Sex Hormones and Inflammatory Cytokines in a Mouse Model of Metoclopramide-Induced Hyperprolactinemia
by Hyeon-Ji Kim, Ji-Woong Jeong, Joo-Yun Kim, Jae-Jung Shim and Jae-Hwan Lee
Nutrients 2024, 16(22), 3889; https://doi.org/10.3390/nu16223889 - 14 Nov 2024
Viewed by 753
Abstract
Background/Objectives: Premenstrual syndrome (PMS), a clinical condition that manifests in the form of various physical and psychological symptoms, occurs periodically during the luteal phase of the menstrual cycle and reduces quality of life. Methods: Here, we conducted in vitro and in vivo experiments [...] Read more.
Background/Objectives: Premenstrual syndrome (PMS), a clinical condition that manifests in the form of various physical and psychological symptoms, occurs periodically during the luteal phase of the menstrual cycle and reduces quality of life. Methods: Here, we conducted in vitro and in vivo experiments to investigate the effects of Lactobacillus helveticus HY7801 (HY7801) on PMS symptoms. Results: Data from the in vitro experiments showed that HY7801 inhibits prolactin secretion by estradiol-induced GH3 cells, as well as the secretion of pro-inflammatory cytokines by LPS-induced Raw 264.7 cells. Additionally, the oral administration of HY7801 (109 colony-forming units/kg/day) to mice with metoclopramide-induced hyperprolactinemia reduced uterine tissue mass and endometrial thickness, both of which were increased excessively in the presence of prolactin. HY7801 also regulated the serum levels of follicle-stimulating hormone and prostaglandin E1/E2, as well as recovering the progesterone/estradiol ratio. HY7801 also downregulated the serum levels of prolactin and pro-inflammatory cytokines such as interleukin (Il)-6, tumor necrosis factor-alpha (Tnf), and IL-1β. Finally, HY7801 reduced the expression of genes encoding inflammatory cytokines (i.e., Tnf and Il-6), cyclooxygenase-2 (Cox-2), and inducible nitric oxide synthase (iNOS) in mice with hyperprolactinemia. Conclusion: In summary, HY7801 may be a functional bacterium that alleviates PMS symptoms by modulating hormones and inflammatory markers. Full article
(This article belongs to the Special Issue Eating Behavior and Women's Health)
Show Figures

Figure 1

Figure 1
<p>Effect of HY7801 on (<b>A</b>) the viability of GH3 cells and (<b>B</b>) the secretion of prolactin by estradiol (E2)-treated GH3 cells. Data are presented as the mean ± SE. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. untreated group; * <span class="html-italic">p</span> &lt; 0.05 vs. E2-treated group. E2: estradiol; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 + E2.</p>
Full article ">Figure 2
<p>Effects of HY7801 on pro-inflammatory cytokines secreted by LPS-induced Raw 264.7 cells. (<b>A</b>) IL-6; (<b>B</b>) TNF-α. Data are presented as the mean ± SE. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. untreated group; *** <span class="html-italic">p</span> &lt; 0.001 vs. LPS-treated group. IL-6: interleukin-6; TNF-α: tumor necrosis factor-alpha; LPS: lipopolysaccharide; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 + LPS.</p>
Full article ">Figure 3
<p>Effect of HY7801 on physiological indicators in mice with MCP-induced hyperprolactinemia. (<b>A</b>) Dietary intake; (<b>B</b>) water intake; (<b>C</b>) change in body weight; (<b>D</b>) mass of uterine tissue; and (<b>E</b>) mass of spleen tissue. Data are presented as the mean ± SE. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. CON group; * <span class="html-italic">p</span> &lt; 0.05 vs. MCP group. CON: non-treatment group; MCP: metoclopramide-induced mice; PFM: prefemin (100 mg/kg/day) + MCP; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 (10<sup>9</sup> CFU/kg/day) + MCP.</p>
Full article ">Figure 4
<p>Histological analysis of uterine tissue from mice with MCP-induced hyperprolactinemia. (<b>A</b>) Hematoxylin and eosin-stained sections; 200× magnification; arrows point to the endometrium. (<b>B</b>) Endometrial thickness. Data are presented as the mean ± SE. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. CON group; *** <span class="html-italic">p</span> &lt; 0.001 vs. MCP group. CON: non-treatment group; MCP: metoclopramide-induced mice; PFM: prefemin (100 mg/kg/day) + MCP; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 (10<sup>9</sup> CFU/kg/day) + MCP.</p>
Full article ">Figure 5
<p>Effect of HY7801 on serum levels of sex hormones in mice with MCP-induced hyperprolactinemia. (<b>A</b>) Prolactin; (<b>B</b>) FSH; and (<b>C</b>) the progesterone/estradiol ratio. Data are presented as the mean ± SE. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. CON group; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. MCP group. FSH: follicle-stimulating hormone; CON: non-treatment group; MCP: metoclopramide-induced mice; PFM: prefemin (100 mg/kg/day) + MCP; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 (10<sup>9</sup> CFU/kg/day) + MCP.</p>
Full article ">Figure 6
<p>Effect of HY7801 on serum levels of pro-inflammatory cytokines and prostaglandin in mice with MCP-induced hyperprolactinemia. (<b>A</b>) IL-6; (<b>B</b>) TNF-α; (<b>C</b>) IL-1β; (<b>D</b>) PGE1; (<b>E</b>) PGE2 levels; and (<b>F</b>) the PGE1/PGE2 ratio. Data are presented as the mean ± SE. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. CON group; * <span class="html-italic">p</span> &lt; 0.05 vs. MCP group. IL-6: interleukin-6; TNF: tumor necrosis factor-alpha; IL-1β: interleukin-1β; PGE1: prostaglandin E1; PGE2: prostaglandin E2; CON: non-treatment group; MCP: metoclopramide-induced mice; PFM: prefemin (100 mg/kg/day) + MCP; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 (10<sup>9</sup> CFU/kg/day) + MCP.</p>
Full article ">Figure 7
<p>Effect of HY7801 on the expression of mRNA encoding inflammation-related genes in uterine tissues from MCP-induced hyperprolactinemia mice. (<b>A</b>) <span class="html-italic">Il-6</span>; (<b>B</b>) <span class="html-italic">Tnf</span>; (<b>C</b>) <span class="html-italic">Cox-2</span>; and (<b>D</b>) <span class="html-italic">iNOS</span>. Data are presented as the mean ± SE. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. CON group; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. MCP group. <span class="html-italic">Il-6</span>: interleukin-6; <span class="html-italic">Tnf</span>: tumor necrosis factor-alpha; <span class="html-italic">Cox-2</span>: cyclooxygenase-2; <span class="html-italic">iNOS</span>: inducible nitric oxide synthase; CON: non-treatment group; MCP: metoclopramide-induced mice; PFM: prefemin (100 mg/kg/day) + MCP; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 (10<sup>9</sup> CFU/kg/day) + MCP.</p>
Full article ">
13 pages, 1124 KiB  
Review
Autophagy Involvement in Non-Neoplastic and Neoplastic Endometrial Pathology: The State of the Art with a Focus on Carcinoma
by Cristina Pizzimenti, Vincenzo Fiorentino, Chiara Ruggeri, Mariausilia Franchina, Alfredo Ercoli, Giovanni Tuccari and Antonio Ieni
Int. J. Mol. Sci. 2024, 25(22), 12118; https://doi.org/10.3390/ijms252212118 - 12 Nov 2024
Viewed by 669
Abstract
Autophagy is a cellular process crucial for maintaining homeostasis by degrading damaged proteins and organelles. It is stimulated in response to stress, recycling nutrients and generating energy for cell survival. In normal endometrium, it suppresses tumorigenesis by preventing toxic accumulation and maintaining cellular [...] Read more.
Autophagy is a cellular process crucial for maintaining homeostasis by degrading damaged proteins and organelles. It is stimulated in response to stress, recycling nutrients and generating energy for cell survival. In normal endometrium, it suppresses tumorigenesis by preventing toxic accumulation and maintaining cellular homeostasis. It is involved in the cyclic remodelling of the endometrium during the menstrual cycle and contributes to decidualisation for successful pregnancy. Such a process is regulated by various signalling pathways, including PI3K/AKT/mTOR, AMPK/mTOR, and p53. Dysregulation of autophagy has been associated with benign conditions like endometriosis and endometrial hyperplasia but also with malignant neoplasms such as endometrial carcinoma. In fact, it has emerged as a crucial player in endometrial carcinoma biology, exhibiting a dual role in both tumour suppression and tumour promotion, providing nutrients during metabolic stress and allowing cancer cell survival. It also regulates cancer stem cells, metastasis and therapy resistance. Targeting autophagy is therefore a promising therapeutic strategy in endometrial carcinoma and potential for overcoming resistance to standard treatments. The aim of this review is to delve into the intricate details of autophagy’s role in endometrial pathology, exploring its mechanisms, signalling pathways and potential therapeutic implications. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Mechanisms of autophagy flux in normal, hyperplastic and dysfunctional uterine pathology (the figure was created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>, accessed on 21 August 2024).</p>
Full article ">Figure 2
<p>The main autophagic-related proteins involved in endometrial carcinoma.</p>
Full article ">
14 pages, 862 KiB  
Article
Is Osteopontin a Reliable Biomarker for Endometriosis?
by Aleksandra Zygula, Kamil Kiecka, Anna Sankiewicz, Mariusz Kuzmicki, Michal Ciebiera, Tadeusz Issat, Wojciech Drygas, Krzysztof Cendrowski, Ewa Gorodkiewicz and Piotr Laudanski
Int. J. Mol. Sci. 2024, 25(22), 11857; https://doi.org/10.3390/ijms252211857 - 5 Nov 2024
Viewed by 799
Abstract
This study aimed to evaluate the concentration of osteopontin in peritoneal fluid and plasma as potential biomarkers for diagnosing endometriosis. Osteopontin levels were measured using surface plasmon resonance imaging (SPRI) biosensors in patients suspected of having endometriosis. Plasma samples were collected from 120 [...] Read more.
This study aimed to evaluate the concentration of osteopontin in peritoneal fluid and plasma as potential biomarkers for diagnosing endometriosis. Osteopontin levels were measured using surface plasmon resonance imaging (SPRI) biosensors in patients suspected of having endometriosis. Plasma samples were collected from 120 patients, and peritoneal fluid was collected from 86 patients. Based on the detection of endometriosis lesions during laparoscopy, participants were divided into a study group (patients with endometriosis) and a control group (patients without endometriosis). The results showed no significant differences in plasma osteopontin levels between women with endometriosis and the control group (19.86 ± 6.72 ng/mL vs. 18.39 ± 4.46 ng/mL, p = 0.15). Similarly, peritoneal fluid osteopontin concentrations did not differ significantly between patients with and without endometriosis (19.04 ± 5.37 ng/mL vs. 17.87 ± 5.13 ng/mL, p = 0.29). Furthermore, osteopontin levels in both plasma and peritoneal fluid were not significantly associated with the stage of endometriosis, the presence of endometrioma, or the menstrual cycle phase. The findings of this study do not support osteopontin concentration as a reliable biomarker for endometriosis. However, further research is necessary to explore osteopontin’s potential role in the disease. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Osteopontin in peritoneal fluid as a predictor of endometriosis AUC = 0.551; ±95% CI (0.430–0.671); <span class="html-italic">p</span> = 0.4108.</p>
Full article ">Figure 2
<p>Osteopontin in plasma as a predictor of endometriosis AUC = 0.582; ±95% CI (0.479–0.686); <span class="html-italic">p</span> = 0.1196.</p>
Full article ">
16 pages, 3272 KiB  
Article
Proof-of-Concept for Long-Term Human Endometrial Epithelial Organoids in Modeling Menstrual Cycle Responses
by Yanyu Jiang, Arturo Reyes Palomares, Patricia Munoz, Ivan Nalvarte, Ganesh Acharya, Jose Inzunza, Mukesh Varshney and Kenny Alexandra Rodriguez-Wallberg
Cells 2024, 13(21), 1811; https://doi.org/10.3390/cells13211811 - 2 Nov 2024
Viewed by 1909
Abstract
Endometrial disorders, such as infertility and endometriosis, significantly impact reproductive health, thus necessitating better models to study endometrial function. Current in vitro models fail to replicate the complexity of the human endometrium throughout the entire menstrual cycle. This study aimed to assess the [...] Read more.
Endometrial disorders, such as infertility and endometriosis, significantly impact reproductive health, thus necessitating better models to study endometrial function. Current in vitro models fail to replicate the complexity of the human endometrium throughout the entire menstrual cycle. This study aimed to assess the physiological response of human endometrial organoids (hEOs) to in vitro hormonal treatments designed to mimic the hormonal fluctuations of the menstrual cycle. Endometrial biopsies from three healthy women were used to develop hEOs, which were treated over 28 days with three hormonal stimulation strategies: (1) estrogen only (E) to mimic the proliferative phase, (2) the addition of progesterone (EP) to simulate the secretory phase, and (3) the further addition of cAMP (EPC) to enhance the secretory functions of hEOs. Gene and protein expression were analyzed using qPCR, IHC, and ELISA. The hEOs exhibited proliferation, gland formation, and appropriate expression of markers such as E-cadherin and Ki67. The hormonal treatments induced significant changes in PR, HSD17B1, PAEP, SPP1, and other genes relevant to endometrial function, closely mirroring in vivo physiological responses. The prominent changes were observed in EPC-treated hEOs (week 4) with significantly high expression of uterine milk components such as glycodelin (PAEP) and osteopontin (SPP1), reflecting mid- to late-secretory phase physiology. This model successfully recapitulates human menstrual cycle dynamics and offers a promising platform for studying endometrial disorders and advancing personalized treatments in gynecology. Full article
(This article belongs to the Section Reproductive Cells and Development)
Show Figures

Figure 1

Figure 1
<p>Endometrial organoids derived from donor biopsy and maintained as glandular structures in long-term culture: (<b>A</b>) anonymized clinical data of donors; (<b>B</b>) extraction of glandular structures and organoid culture establishment; (<b>C</b>) hormone treatment conditions of endometrial organoids for 28 days period; (<b>D</b>) representative bright field images of endometrial organoids from Donor 1 at different days in long-term culture in spinning bioreactor. Scale bar 100 µm for image panel in (<b>D</b>).</p>
Full article ">Figure 2
<p>Human endometrium organoids respond to estrogen treatment: (<b>A</b>) treatment plan and violin plots of gene expression changes in hEOs during 28 days with estrogen (E2) treatment measured by quantitative polymerase chain reaction (qPCR); (<b>B</b>) representative immunofluorescent images showing, Ki67, PGR (PR), E-cadherin (E-Cad), and PAEP expression through 28-day estrogen treatment in hEOs from donor 1 (cyan), donor 2 (red), and donor 3 (green). Results illustrated in (<b>A</b>) represent mean ± SD from three independent biological replicates (<span class="html-italic">n</span> = 3 donors), each with three experimental replicates (<span class="html-italic">n</span> = 9 total) and analyzed by one-way ANOVA with Tukey’s multiple-comparison test and post hoc correction, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001; scale bar in (<b>B</b>): 50 µm.</p>
Full article ">Figure 3
<p>Sequential estrogen and progesterone treatment mimics the secretory phase in human endometrial organoids: (<b>A</b>) treatment plan and violin plots of gene expression changes in hEOs during 28 days with sequential progesterone (P4) treatment following estrogen (E2) day 14 onwards, measured by quantitative polymerase chain reaction (qPCR); (<b>B</b>) representative immunofluorescent images showing ESR1 (ER), PGR (PR), and PAEP expression through 28-day treatment in hEOs from donor 1 (cyan), donor 2 (red), and donor 3 (green). Results illustrated in (<b>A</b>) represent mean ± SD from three independent biological replicates (<span class="html-italic">n</span> = 3 donors), each with three experimental replicates (<span class="html-italic">n</span> = 9 total), and analyzed by one-way ANOVA with Tukey’s multiple-comparison test and post hoc correction, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001; scale bar in (<b>B</b>): 50 µm.</p>
Full article ">Figure 4
<p>cAMP enhances progesterone-induced differentiation of human endometrial organoids: (<b>A</b>) treatment plan and violin plots of gene expression changes in hEOs during 28 days with sequential progesterone (P4) and cAMP treatment following estrogen (E2) treatment from day 14 onwards in hEOs measured by quantitative polymerase chain reaction (qPCR); (<b>B</b>) representative immunofluorescent images showing ESR1 (ER), PGR (PR), and PAEP expression at day 21 and 28 through 28-day treatment in hEOs from donor 1 (cyan), donor 2 (red), and donor 3 (green). Results illustrated in (<b>A</b>) represent mean ± SD from three independent biological replicates (<span class="html-italic">n</span> = 3 donors), each with three experimental replicates (<span class="html-italic">n</span> = 9 total), and analyzed by one-way ANOVA with Tukey’s multiple-comparison test and post hoc correction, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001; scale bar in (<b>B</b>): 50 µm.</p>
Full article ">Figure 5
<p>Human endometrium organoids treated with sequential estrogen and progesterone with cAMP induce the secretory phase: (<b>A</b>) treatment plan and bright field images of endometrial organoids cultured at 18 days in vitro with different hormone treatments in hEOs; (<b>B</b>) heatmap of gene expression changes at day 21 in hEOs measured by quantitative polymerase chain reaction (qPCR); (<b>C</b>) violin plot of ELISA for secreted PAEP (left) and SPP1 (right) from culture supernatant with different hormone treatments at day 21 in hEOs. Results illustrated in (<b>B</b>) represent mean ± SD (<span class="html-italic">n</span> = 3) and were analyzed by two-way ANOVA with Sidak multiple comparisons; the results in C represent mean ± SD from three independent biological replicates (<span class="html-italic">n</span> = 3 donors), each with three experimental replicates (<span class="html-italic">n</span> = 9 total), and analyzed by one-way ANOVA with Tukey’s multiple-comparison test and post hoc correction, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001; scale bar in (A): 50 µm.</p>
Full article ">
Back to TopTop