140 results on '"Giudice, P."'
Search Results
2. Safety and efficacy of immune checkpoint inhibitors in advanced penile cancer: report from the Global Society of Rare Genitourinary Tumors.
- Author
-
El Zarif, Talal, Nassar, Amin, Pond, Gregory, Zhuang, Tony, Master, Viraj, Nazha, Bassel, Niglio, Scot, Simon, Nicholas, Hahn, Andrew, Pettaway, Curtis, Tu, Shi-Ming, Abdel-Wahab, Noha, Velev, Maud, Flippot, Ronan, Buti, Sebastiano, Maruzzo, Marco, Mittra, Arjun, Gheeya, Jinesh, Yang, Yuanquan, Rodriguez, Pablo, Castellano, Daniel, de Velasco, Guillermo, Roviello, Giandomenico, Antonuzzo, Lorenzo, McKay, Rana, Vincenzi, Bruno, Cortellini, Alessio, Hui, Gavin, Drakaki, Alexandra, Glover, Michael, Khaki, Ali, El-Am, Edward, Adra, Nabil, Mouhieddine, Tarek, Patel, Vaibhav, Piedra, Aida, Gernone, Angela, Davis, Nancy, Matthews, Harrison, Harrison, Michael, Kanesvaran, Ravindran, Giudice, Giulia, Barata, Pedro, Farolfi, Alberto, Lee, Jae, Milowsky, Matthew, Stahlfeld, Charlotte, Appleman, Leonard, Kim, Joseph, Freeman, Dory, Choueiri, Toni, Spiess, Philippe, Necchi, Andrea, Apolo, Andrea, and Sonpavde, Guru
- Subjects
Male ,Humans ,Middle Aged ,Aged ,Nivolumab ,Immune Checkpoint Inhibitors ,Penile Neoplasms ,Antineoplastic Agents ,Immunological ,Retrospective Studies ,Carcinoma ,Squamous Cell ,Antineoplastic Combined Chemotherapy Protocols - Abstract
BACKGROUND: Treatment options for penile squamous cell carcinoma are limited. We sought to investigate clinical outcomes and safety profiles of patients with penile squamous cell carcinoma receiving immune checkpoint inhibitors. METHODS: This retrospective study included patients with locally advanced or metastatic penile squamous cell carcinoma receiving immune checkpoint inhibitors between 2015 and 2022 across 24 centers in the United States, Europe, and Asia. Overall survival and progression-free survival were estimated using the Kaplan-Meier method. Objective response rates were determined per Response Evaluation Criteria in Solid Tumours 1.1 criteria. Treatment-related adverse events were graded per the Common Terminology Criteria for Adverse Events, version 5.0. Two-sided statistical tests were used for comparisons. RESULTS: Among 92 patients, 8 (8.7%) were Asian, 6 (6.5%) were Black, and 24 (29%) were Hispanic and/or Latinx. Median (interquartile range) age was 62 (53-70) years. In all, 83 (90%) had metastatic penile squamous cell carcinoma, and 74 (80%) had received at least second-line treatment. Most patients received pembrolizumab monotherapy (n = 26 [28%]), combination nivolumab-ipilimumab with or without multitargeted tyrosine kinase inhibitors (n = 23 [25%]), or nivolumab (n = 16 [17%]) or cemiplimab (n = 15 [16%]) monotherapies. Median overall and progression-free survival were 9.8 months (95% confidence interval = 7.7 to 12.8 months) and 3.2 months (95% confidence interval = 2.5 to 4.2 months), respectively. The objective response rate was 13% (n = 11/85) in the overall cohort and 35% (n = 7/20) in patients with lymph node-only metastases. Visceral metastases, Eastern Cooperative Oncology Group (ECOG) performance status of 1 or higher, and a higher neutrophil/lymphocyte ratio were associated with worse overall survival. Treatment-related adverse events occurred in 27 (29%) patients, and 9.8% (n = 9) of the events were grade 3 or higher. CONCLUSIONS: Immune checkpoint inhibitors are active in a subset of patients with penile squamous cell carcinoma. Future translational studies are warranted to identify patients more likely to derive clinical benefit from immune checkpoint inhibitors.
- Published
- 2023
3. Endometriosis in the era of precision medicine and impact on sexual and reproductive health across the lifespan and in diverse populations
- Author
-
Giudice, Linda C, Oskotsky, Tomiko T, Falako, Simileoluwa, Opoku‐Anane, Jessica, and Sirota, Marina
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Patient Safety ,Infertility ,Clinical Research ,Pain Research ,Endometriosis ,Prevention ,Contraception/Reproduction ,Chronic Pain ,Genetics ,2.1 Biological and endogenous factors ,Aetiology ,Reproductive health and childbirth ,Good Health and Well Being ,Adolescent ,Humans ,Female ,Aged ,80 and over ,Precision Medicine ,Longevity ,Artificial Intelligence ,Quality of Life ,Reproductive Health ,access ,biomarkers ,diagnosis ,diversity ,endometriosis ,equity ,health disparities ,precision medicine ,therapies ,Biochemistry and Cell Biology ,Physiology ,Medical Physiology ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medical physiology - Abstract
Endometriosis is a common estrogen-dependent disorder wherein uterine lining tissue (endometrium) is found mainly in the pelvis where it causes inflammation, chronic pelvic pain, pain with intercourse and menses, and infertility. Recent evidence also supports a systemic inflammatory component that underlies associated co-morbidities, e.g., migraines and cardiovascular and autoimmune diseases. Genetics and environment contribute significantly to disease risk, and with the explosion of omics technologies, underlying mechanisms of symptoms are increasingly being elucidated, although novel and effective therapeutics for pain and infertility have lagged behind these advances. Moreover, there are stark disparities in diagnosis, access to care, and treatment among persons of color and transgender/nonbinary identity, socioeconomically disadvantaged populations, and adolescents, and a disturbing low awareness among health care providers, policymakers, and the lay public about endometriosis, which, if left undiagnosed and under-treated can lead to significant fibrosis, infertility, depression, and markedly diminished quality of life. This review summarizes endometriosis epidemiology, compelling evidence for its pathogenesis, mechanisms underlying its pathophysiology in the age of precision medicine, recent biomarker discovery, novel therapeutic approaches, and issues around reproductive justice for marginalized populations with this disorder spanning the past 100 years. As we enter the next revolution in health care and biomedical research, with rich molecular and clinical datasets, single-cell omics, and population-level data, endometriosis is well positioned to benefit from data-driven research leveraging computational and artificial intelligence approaches integrating data and predicting disease risk, diagnosis, response to medical and surgical therapies, and prognosis for recurrence.
- Published
- 2023
4. Pregnancy outcomes and vaccine effectiveness during the period of omicron as the variant of concern, INTERCOVID-2022: a multinational, observational study
- Author
-
Villar, Jose, Conti, Constanza P Soto, Gunier, Robert B, Ariff, Shabina, Craik, Rachel, Cavoretto, Paolo I, Rauch, Stephen, Gandino, Serena, Nieto, Ricardo, Winsey, Adele, Menis, Camilla, Rodriguez, Gabriel B, Savasi, Valeria, Tug, Niyazi, Deantoni, Sonia, Fabre, Marta, de Tejada, Begoña Martinez, Rodriguez-Sibaja, Maria Jose, Livio, Stefania, Napolitano, Raffaele, Maiz, Nerea, Sobrero, Helena, Peterson, Ashley, Deruelle, Philippe, Giudice, Carolina, Teji, Jagjit S, Casale, Roberto A, Salomon, Laurent J, Prefumo, Federico, Ismail, Leila Cheikh, Gravett, Michael G, Vale, Marynéa, Hernández, Valeria, Sentilhes, Loïc, Easter, Sarah R, Capelli, Carola, Marler, Emily, Cáceres, Daniela M, Crespo, Guadalupe Albornoz, Ernawati, Ernawati, Lipschuetz, Michal, Takahashi, Ken, Vecchiarelli, Carmen, Hubka, Teresa, Ikenoue, Satoru, Tavchioska, Gabriela, Bako, Babagana, Ayede, Adejumoke I, Eskenazi, Brenda, Thornton, Jim G, Bhutta, Zulfiqar A, Kennedy, Stephen H, Papageorghiou, Aris T, and Consortium, INTERCOVID-2022 International
- Subjects
Prevention ,Pediatric ,Lung ,Clinical Research ,Immunization ,Clinical Trials and Supportive Activities ,Vaccine Related ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,Reproductive health and childbirth ,Good Health and Well Being ,Pregnancy ,Infant ,Newborn ,Humans ,Female ,Male ,Pregnancy Outcome ,Vaccine Efficacy ,COVID-19 ,SARS-CoV-2 ,COVID-19 Testing ,Prospective Studies ,Mothers ,INTERCOVID-2022 International Consortium ,Medical and Health Sciences ,General & Internal Medicine - Abstract
BackgroundIn 2021, we showed an increased risk associated with COVID-19 in pregnancy. Since then, the SARS-CoV-2 virus has undergone genetic mutations. We aimed to examine the effects on maternal and perinatal outcomes of COVID-19 during pregnancy, and evaluate vaccine effectiveness, when omicron (B.1.1.529) was the variant of concern.MethodsINTERCOVID-2022 is a large, prospective, observational study, involving 41 hospitals across 18 countries. Each woman with real-time PCR or rapid test, laboratory-confirmed COVID-19 in pregnancy was compared with two unmatched women without a COVID-19 diagnosis who were recruited concomitantly and consecutively in pregnancy or at delivery. Mother and neonate dyads were followed until hospital discharge. Primary outcomes were maternal morbidity and mortality index (MMMI), severe neonatal morbidity index (SNMI), and severe perinatal morbidity and mortality index (SPMMI). Vaccine effectiveness was estimated, adjusted by maternal risk profile.FindingsWe enrolled 4618 pregnant women from Nov 27, 2021 (the day after WHO declared omicron a variant of concern), to June 30, 2022: 1545 (33%) women had a COVID-19 diagnosis (median gestation 36·7 weeks [IQR 29·0-38·9]) and 3073 (67%) women, with similar demographic characteristics, did not have a COVID-19 diagnosis. Overall, women with a diagnosis had an increased risk for MMMI (relative risk [RR] 1·16 [95% CI 1·03-1·31]) and SPMMI (RR 1·21 [95% CI 1·00-1·46]). Women with a diagnosis, compared with those without a diagnosis, also had increased risks of SNMI (RR 1·23 [95% CI 0·88-1·71]), although the lower bounds of the 95% CI crossed unity. Unvaccinated women with a COVID-19 diagnosis had a greater risk of MMMI (RR 1·36 [95% CI 1·12-1·65]). Severe COVID-19 symptoms in the total sample increased the risk of severe maternal complications (RR 2·51 [95% CI 1·84-3·43]), perinatal complications (RR 1·84 [95% CI 1·02-3·34]), and referral, intensive care unit (ICU) admission, or death (RR 11·83 [95% CI 6·67-20·97]). Severe COVID-19 symptoms in unvaccinated women increased the risk of MMMI (RR 2·88 [95% CI 2·02-4·12]) and referral, ICU admission, or death (RR 20·82 [95% CI 10·44-41·54]). 2886 (63%) of 4618 total participants had at least a single dose of any vaccine, and 2476 (54%) of 4618 had either complete or booster doses. Vaccine effectiveness (all vaccines combined) for severe complications of COVID-19 for all women with a complete regimen was 48% (95% CI 22-65) and 76% (47-89) after a booster dose. For women with a COVID-19 diagnosis, vaccine effectiveness of all vaccines combined for women with a complete regimen was 74% (95% CI 48-87) and 91% (65-98) after a booster dose.InterpretationCOVID-19 in pregnancy, during the first 6 months of omicron as the variant of concern, was associated with increased risk of severe maternal morbidity and mortality, especially among symptomatic and unvaccinated women. Women with complete or boosted vaccine doses had reduced risk for severe symptoms, complications, and death. Vaccination coverage among pregnant women remains a priority.FundingNone.
- Published
- 2023
5. Time for global health policy and research leaders to prioritize endometriosis
- Author
-
Giudice, Linda C, Horne, Andrew W, and Missmer, Stacey A
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Health Sciences ,Clinical Research ,Infertility ,Contraception/Reproduction ,Pain Research ,Endometriosis ,Chronic Pain ,Digestive Diseases ,Good Health and Well Being ,Female ,Humans ,Global Health ,Health Policy - Abstract
Endometriosis is an incurable, under-diagnosed, systemic inflammatory disease affecting millions world-wide. Common symptoms include life-impacting pain, gastrointestinal/urinary symptoms, excessive fatigue, and infertility. Global public health policies are urgently needed to promote awareness, implement multidisciplinary care, and fund research for aetiology, biomarker discovery, and effective therapies for symptoms associated with endometriosis.
- Published
- 2023
6. Global endometrial DNA methylation analysis reveals insights into mQTL regulation and associated endometriosis disease risk and endometrial function
- Author
-
Mortlock, Sally, Houshdaran, Sahar, Kosti, Idit, Rahmioglu, Nilufer, Nezhat, Camran, Vitonis, Allison F, Andrews, Shan V, Grosjean, Parker, Paranjpe, Manish, Horne, Andrew W, Jacoby, Alison, Lager, Jeannette, Opoku-Anane, Jessica, Vo, Kim Chi, Manvelyan, Evelina, Sen, Sushmita, Ghukasyan, Zhanna, Collins, Frances, Santamaria, Xavier, Saunders, Philippa, Kober, Kord, McRae, Allan F, Terry, Kathryn L, Vallvé-Juanico, Júlia, Becker, Christian, Rogers, Peter AW, Irwin, Juan C, Zondervan, Krina, Montgomery, Grant W, Missmer, Stacey, Sirota, Marina, and Giudice, Linda
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Genetics ,Biological Sciences ,Pain Research ,Infertility ,Contraception/Reproduction ,Human Genome ,Biotechnology ,Endometriosis ,Aetiology ,2.1 Biological and endogenous factors ,Reproductive health and childbirth ,Good Health and Well Being ,Female ,Humans ,DNA Methylation ,Pain ,Embryo Implantation ,Biological sciences ,Biomedical and clinical sciences - Abstract
Endometriosis is a leading cause of pain and infertility affecting millions of women globally. Herein, we characterize variation in DNA methylation (DNAm) and its association with menstrual cycle phase, endometriosis, and genetic variants through analysis of genotype data and methylation in endometrial samples from 984 deeply-phenotyped participants. We estimate that 15.4% of the variation in endometriosis is captured by DNAm and identify significant differences in DNAm profiles associated with stage III/IV endometriosis, endometriosis sub-phenotypes and menstrual cycle phase, including opening of the window for embryo implantation. Menstrual cycle phase was a major source of DNAm variation suggesting cellular and hormonally-driven changes across the cycle can regulate genes and pathways responsible for endometrial physiology and function. DNAm quantitative trait locus (mQTL) analysis identified 118,185 independent cis-mQTLs including 51 associated with risk of endometriosis, highlighting candidate genes contributing to disease risk. Our work provides functional evidence for epigenetic targets contributing to endometriosis risk and pathogenesis. Data generated serve as a valuable resource for understanding tissue-specific effects of methylation on endometrial biology in health and disease.
- Published
- 2023
7. Deep immunophenotyping reveals endometriosis is marked by dysregulation of the mononuclear phagocytic system in endometrium and peripheral blood
- Author
-
Vallvé-Juanico, Júlia, George, Ashley F, Sen, Sushmita, Thomas, Reuben, Shin, Min-Gyoung, Kushnoor, Divyashree, Vásquez, Joshua J, Vo, Kim Chi, Irwin, Juan C, Roan, Nadia R, Combes, Alexis J, and Giudice, Linda C
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Endometriosis ,Contraception/Reproduction ,Pain Research ,Clinical Research ,Underpinning research ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Aetiology ,Inflammatory and immune system ,Reproductive health and childbirth ,Case-Control Studies ,Endometrium ,Female ,Humans ,Immunophenotyping ,Inflammation ,Macrophages ,Monocytes ,Mononuclear phagocytes ,SIRP alpha ,Innate immune ,CyTOF ,Biomarker ,SIRPα ,Medical and Health Sciences ,General & Internal Medicine ,Biomedical and clinical sciences ,Health sciences - Abstract
BackgroundEndometriosis is a chronic, estrogen-dependent disorder where inflammation contributes to disease-associated symptoms of pelvic pain and infertility. Immune dysfunction includes insufficient immune lesion clearance, a pro-inflammatory endometrial environment, and systemic inflammation. Comprehensive understanding of endometriosis immune pathophysiology in different hormonal milieu and disease severity has been hampered by limited direct characterization of immune populations in endometrium, blood, and lesions. Simultaneous deep phenotyping at single-cell resolution of complex tissues has transformed our understanding of the immune system and its role in many diseases. Herein, we report mass cytometry and high dimensional analyses to study immune cell phenotypes, abundance, activation states, and functions in endometrium and blood of women with and without endometriosis in different cycle phases and disease stages.MethodsA case-control study was designed. Endometrial biopsies and blood (n = 60 total) were obtained from women with (n = 20, n = 17, respectively) and without (n = 14, n = 9) endometriosis in the proliferative and secretory cycle phases of the menstrual cycle. Two mass cytometry panels were designed: one broad panel and one specific for mononuclear phagocytic cells (MPC), and all samples were multiplexed to characterize both endometrium and blood immune composition at unprecedented resolution. We combined supervised and unsupervised analyses to finely define the immune cell subsets with an emphasis on MPC. Then, association between cell types, protein expression, disease status, and cycle phase were performed.ResultsThe broad panel highlighted a significant modification of MPC in endometriosis; thus, they were studied in detail with an MPC-focused panel. Endometrial CD91+ macrophages overexpressed SIRPα (phagocytosis inhibitor) and CD64 (associated with inflammation) in endometriosis, and they were more abundant in mild versus severe disease. In blood, classical and intermediate monocytes were less abundant in endometriosis, whereas plasmacytoid dendritic cells and non-classical monocytes were more abundant. Non-classical monocytes were higher in severe versus mild disease.ConclusionsA greater inflammatory phenotype and decreased phagocytic capacity of endometrial macrophages in endometriosis are consistent with defective clearance of endometrial cells shed during menses and in tissue homeostasis, with implications in endometriosis pathogenesis and pathophysiology. Different proportions of monocytes and plasmacytoid dendritic cells in blood from endometriosis suggest systemically aberrant functionality of the myeloid system opening new venues for the study of biomarkers and therapies for endometriosis.
- Published
- 2022
8. The FIGO Ovulatory Disorders Classification System†
- Author
-
Munro, Malcolm G, Balen, Adam H, Cho, SiHyun, Critchley, Hilary OD, Díaz, Ivonne, Ferriani, Rui, Henry, Laurie, Mocanu, Edgar, van der Spuy, Zephne M, Acharya, Ganesh, Adonakis, Georgios, Ahsan, Sadiah, AIhaidari, Taghreed, Asatiani, Tengiz, Azziz, Ricardo, Balen, Adam, Bedard, Michela, Blake, Jennifer, Chamy, Veronica, Cheong, Ying, Cheung, Vincent YT, Cho, Si Hyun, Critchley, Hilary, da Silva, Jose Teixeira, Diaz, Ivonne, Duncan, Colin, Ekersley, Amelie, Epifanio-Malpassii, Roberto, Famuyide, Abimbola, Giudice, Linda, Gurevich, Maargarita, Harlow, Sioban, Hart, Roger, Heikinheimo, Oskari, Heylen, Sulaiman, Kennedy, Richard, Klepchuckova, Anna, Krepelka, Petr, le Roux, Paul, Levchenko, Kateryna, Loutradis, Dimitrios, Marsh, Erica, Martins, Noni, Mathur, Raj, Matsaseng, Thabo, Miguelote, Rui, Munro, Malcolm, Ngoga, Eugene, Nisolle, Michelle, Norman, Robert, Ono, Masanori, Pintiaux, Axelle, Pristauz-Telsnigg, Gunda, Ramasauskaite, Diana, Ravn, Pernille, Reis, Jose, Roos, Peter, Rozic, Irena, Scarella, Anibal, Sharai, Katsiaryna, Shibut, Alena, Sierra, Sony, Steiner, Anne, Stoop, Dominic, Toth, Bettina, Van Der Spuy, Zephne, Williams, Saskia, Wise, Lauren, Yazdani, Anusch, Zhaunova, Liudmila, Zunckel, Meggan, and Zwane, Karabo
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Contraception/Reproduction ,Clinical Research ,Reproductive health and childbirth ,Good Health and Well Being ,Endocrinology ,Female ,Gynecology ,Humans ,Polycystic Ovary Syndrome ,Pregnancy ,Uterine Diseases ,anovulation ,ovulatory disorders ,ovulatory disorders classification ,ovulatory dysfunction ,FIGO Committee on Menstrual Disorders and Related Health Impacts ,and FIGO Committee on Reproductive Medicine ,Endocrinology ,and Infertility ,Medical and Health Sciences ,Studies in Human Society ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Abstract
Ovulatory disorders are common causes of amenorrhea, abnormal uterine bleeding and infertility and are frequent manifestations of polycystic ovary syndrome (PCOS). There are many potential causes and contributors to ovulatory dysfunction that challenge clinicians, trainees, educators, and those who perform basic, translational, clinical and epidemiological research. Similarly, therapeutic approaches to ovulatory dysfunction potentially involve a spectrum of lifestyle, psychological, medical and procedural interventions. Collaborative research, effective education and consistent clinical care remain challenged by the absence of a consensus comprehensive system for classification of these disorders. The existing and complex system, attributed to the World Health Organization (WHO), was developed more than three decades ago and did not consider more than 30 years of research into these disorders in addition to technical advances in imaging and endocrinology. This article describes the development of a new classification of ovulatory disorders performed under the aegis of the International Federation of Gynecology and Obstetrics (FIGO) and conducted using a rigorously applied Delphi process. The stakeholder organizations and individuals who participated in this process comprised specialty journals, experts at large, national, specialty obstetrical and gynecological societies, and informed lay representatives. After two face-to-face meetings and five Delphi rounds, the result is a three-level multi-tiered system. The system is applied after a preliminary assessment identifies the presence of an ovulatory disorder. The primary level of the system is based on an anatomic model (Hypothalamus, Pituitary, Ovary) that is completed with a separate category for PCOS. This core component of the system is easily remembered using the acronym HyPO-P. Each anatomic category is stratified in the second layer of the system to provide granularity for investigators, clinicians and trainees using the 'GAIN-FIT-PIE' mnemonic (Genetic, Autoimmune, Iatrogenic, Neoplasm; Functional, Infectious and Inflammatory, Trauma and Vascular; Physiological, Idiopathic, Endocrine). The tertiary level allows for specific diagnostic entities. It is anticipated that, if widely adopted, this system will facilitate education, clinical care and the design and interpretation of research in a fashion that better informs progress in this field. Integral to the deployment of this system is a periodic process of reevaluation and appropriate revision, reflecting an improved understanding of this collection of disorders.
- Published
- 2022
9. Transcriptomic analysis supports collective endometrial cell migration in the pathogenesis of adenomyosis
- Author
-
Zhai, Junyu, Li, Shang, Sen, Sushmita, Vallvé-Juanico, Júlia, Irwin, Juan C, Vo, Kim Chi, Wan, Jipeng, Du, Yanzhi, Chen, Zi-Jiang, and Giudice, Linda C
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Contraception/Reproduction ,Genetics ,Rare Diseases ,Pain Research ,2.1 Biological and endogenous factors ,Aetiology ,Adenomyosis ,Cell Movement ,Endometriosis ,Endometrium ,Female ,Humans ,Inflammation ,Prolactin ,Transcriptome ,gamma-Aminobutyric Acid ,Collective cell migration ,ECM remodeling ,GABA ,RNA-seq ,Paediatrics and Reproductive Medicine ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Abstract
Research questionAdenomyosis is a common uterine disorder of uncertain causes. Can transcriptomic analyses of the endometrium and myometrium reveal potential mechanisms underlying adenomyosis pathogenesis?DesignTranscriptomic profiles of eutopic endometrium and myometrium from women with and without diffuse adenomyosis and with symptomatic FIGO type 2-5 fibroids in the proliferative phase of the menstrual cycle were assessed using RNA sequencing and bioinformatic analysis. Differentially expressed genes (DEG) and potential pathways were validated by quantitative reverse transcription polymerase chain reaction, immunoblotting and Masson staining, using additional clinical samples.ResultsTop biological processes in the endometrium of women with versus without adenomyosis, enriched from DEG, comprised inflammation, extracellular matrix (ECM) organization, collagen degradation and hyaluronan synthesis, which are key in cell migration and cell movement. Top biological processes enriched from DEG in the myometrium of women with versus without adenomyosis revealed ECM organization dysfunction, abnormal sensory pain perception and gamma aminobutyric acid (GABA) synaptic transmission. Dysregulation of prolactin signalling was also enriched in eutopic endometrium and in the myometrium of women with adenomyosis.ConclusionsOverall, our results support the invasive endometrium theory in the pathogenesis of adenomyosis, in which inflammation induces ECM remodelling resulting in a track for subsequent endometrial collective cell migration and onset of adenomyosis. Moreover, abnormal myometrial GABA synaptic transmission may contribute to dysmenorrhoea in women with adenomyosis and is a possible target for novel therapeutic development. Prolactin signalling abnormalities may serve as another opportunity for therapeutic intervention.
- Published
- 2022
10. Nutritional interventions to ameliorate the effect of endocrine disruptors on human reproductive health: A semi‐structured review from FIGO
- Author
-
Corbett, Gillian A, Lee, Sadhbh, Woodruff, Tracey J, Hanson, Mark, Hod, Moshe, Charlesworth, Anne Marie, Giudice, Linda, Conry, Jeanne, McAuliffe, Fionnuala M, and Exposures, International Federation of Gynecology and Obstetrics Committee on Impact of Pregnancy on Long‐term Health and the FIGO Committee on Climate Change and Toxic Environmental
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Estrogen ,Prevention ,Clinical Research ,Clinical Trials and Supportive Activities ,Nutrition ,3.3 Nutrition and chemoprevention ,Prevention of disease and conditions ,and promotion of well-being ,Reproductive health and childbirth ,Good Health and Well Being ,Endocrine Disruptors ,Female ,Humans ,Pregnancy ,Reproductive Health ,Vitamins ,endocrine disruptors ,lifestyle interventions ,nutritional interventions ,perinatal morbidity ,reproductive outcomes ,International Federation of Gynecology and Obstetrics (FIGO) Committee on Impact of Pregnancy on Long-term Health and the FIGO Committee on Climate Change and Toxic Environmental Exposures ,Paediatrics and Reproductive Medicine ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Abstract
BackgroundEndocrine disrupting chemicals have harmful effects on reproductive, perinatal, and obstetric outcomes.ObjectiveTo analyze the evidence on nutritional interventions to reduce the negative effects of endocrine disruptors on reproductive, perinatal, and obstetric outcomes.Search strategyA search of MEDLINE (PubMed), Allied Health Literature (CINAHL), EMBASE, Web of Science, and the Cochrane Database was conducted from inception to May 2021.Selection criteriaExperimental studies on human populations.Data collection and analysisData were collected from eligible studies. Risk of bias assessment was completed using the Cochrane risk of bias tool and the ROBINS-I Tool.ResultsDatabase searches yielded 15 362 articles. Removing 11 181 duplicates, 4181 articles underwent abstract screening, 26 articles were eligible for full manuscript review, and 16 met full inclusion criteria. Several interventions were found to be effective in reducing exposure to endocrine disruptors: avoidance of plastic containers, bottles, and packaging; avoidance of canned food/beverages; consumption of fresh and organic food; avoidance of fast/processed foods; and supplementation with vitamin C, iodine, and folic acid. There were some interventional studies examining therapies to improve clinical outcomes related to endocrine disruptors.ConclusionDietary alterations can reduce exposure to endocrine disruptors, with limited data on interventions to improve endocrine-disruptor-related clinical outcomes. This review provides useful instruction to women, their families, healthcare providers, and regulatory bodies.
- Published
- 2022
11. Whole-Tissue Deconvolution and scRNAseq Analysis Identify Altered Endometrial Cellular Compositions and Functionality Associated With Endometriosis
- Author
-
Bunis, Daniel G, Wang, Wanxin, Vallvé-Juanico, Júlia, Houshdaran, Sahar, Sen, Sushmita, Soltane, Isam Ben, Kosti, Idit, Vo, Kim Chi, Irwin, Juan C, Giudice, Linda C, and Sirota, Marina
- Subjects
Cancer ,Pain Research ,Endometriosis ,Clinical Research ,Contraception/Reproduction ,Uterine Cancer ,Infertility ,Aetiology ,Underpinning research ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Inflammatory and immune system ,Reproductive health and childbirth ,Endometrium ,Female ,Humans ,RNA-Seq ,Single-Cell Analysis ,endometriosis ,deconvolution ,bulk tissue transcriptomics ,single-cell analysis ,eutopic endometrium ,Immunology ,Medical Microbiology - Abstract
The uterine lining (endometrium) exhibits a pro-inflammatory phenotype in women with endometriosis, resulting in pain, infertility, and poor pregnancy outcomes. The full complement of cell types contributing to this phenotype has yet to be identified, as most studies have focused on bulk tissue or select cell populations. Herein, through integrating whole-tissue deconvolution and single-cell RNAseq, we comprehensively characterized immune and nonimmune cell types in the endometrium of women with or without disease and their dynamic changes across the menstrual cycle. We designed metrics to evaluate specificity of deconvolution signatures that resulted in single-cell identification of 13 novel signatures for immune cell subtypes in healthy endometrium. Guided by statistical metrics, we identified contributions of endometrial epithelial, endothelial, plasmacytoid dendritic cells, classical dendritic cells, monocytes, macrophages, and granulocytes to the endometrial pro-inflammatory phenotype, underscoring roles for nonimmune as well as immune cells to the dysfunctionality of this tissue.
- Published
- 2022
12. Climate change, women’s health, and the role of obstetricians and gynecologists in leadership
- Author
-
Giudice, Linda C, Llamas‐Clark, Erlidia F, DeNicola, Nathaniel, Pandipati, Santosh, Zlatnik, Marya G, Decena, Ditas Cristina D, Woodruff, Tracey J, Conry, Jeanne A, and Exposures, the FIGO Committee on Climate Change and Toxic Environmental
- Subjects
Global Warming Climate Change ,Climate-Related Exposures and Conditions ,Climate Change ,Reproductive health and childbirth ,Climate Action ,Aged ,Air Pollution ,Animals ,Female ,Humans ,Leadership ,Pregnancy ,Public Health ,Women's Health ,advocacy ,climate change ,education ,environment ,reproduction ,women's health ,FIGO Committee on Climate Change and Toxic Environmental Exposures ,Paediatrics and Reproductive Medicine ,Obstetrics & Reproductive Medicine - Abstract
Climate change is one of the major global health threats to the world's population. It is brought on by global warming due in large part to increasing levels of greenhouse gases resulting from human activity, including burning fossil fuels (carbon dioxide), animal husbandry (methane from manure), industry emissions (ozone, nitrogen oxides, sulfur dioxide), vehicle/factory exhaust, and chlorofluorocarbon aerosols that trap extra heat in the earth's atmosphere. Resulting extremes of weather give rise to wildfires, air pollution, changes in ecology, and floods. These in turn result in displacement of populations, family disruption, violence, and major impacts on water quality and availability, food security, public health and economic infrastructures, and limited abilities for civil society to maintain citizen safety. Climate change also has direct impacts on human health and well-being. Particularly vulnerable populations are affected, including women, pregnant women, children, the disabled, and the elderly, who comprise the majority of the poor globally. Additionally, the effects of climate change disproportionally affect disadvantaged communities, including low income and communities of color, and lower-income countries that are at highest risk of adverse impacts when disasters occur due to inequitable distribution of resources and their socioeconomic status. The climate crisis is tilting the risk balance unfavorably for women's sexual and reproductive health and rights as well as newborn and child health. Obstetrician/gynecologists have the unique opportunity to raise awareness, educate, and advocate for mitigation strategies to reverse climate change affecting our patients and their families. This article puts climate change in the context of women's reproductive health as a public health issue, a social justice issue, a human rights issue, an economic issue, a political issue, and a gender issue that needs our attention now for the health and well-being of this and future generations. FIGO joins a broad coalition of international researchers and the medical community in stating that the current climate crisis presents an imminent health risk to pregnant people, developing fetuses, and reproductive health, and recognizing that we need society-wide solutions, government policies, and global cooperation to address and reduce contributors, including fossil fuel production, to climate change.
- Published
- 2021
13. Biomarkers in Prostate Cancer Diagnosis: From Current Knowledge to the Role of Metabolomics and Exosomes.
- Author
-
Salciccia, Stefano, Capriotti, Anna Laura, Laganà, Aldo, Fais, Stefano, Logozzi, Mariantonia, De Berardinis, Ettore, Busetto, Gian Maria, Di Pierro, Giovanni Battista, Ricciuti, Gian Piero, Del Giudice, Francesco, Sciarra, Alessandro, Carroll, Peter R, Cooperberg, Matthew R, Sciarra, Beatrice, and Maggi, Martina
- Subjects
biomarkers ,early diagnosis ,exosomes ,metabolomics ,prostate cancer ,Animals ,Biomarkers ,Tumor ,Exosomes ,Humans ,Male ,Metabolome ,Prostatic Neoplasms ,Biomarkers ,Tumor ,Other Chemical Sciences ,Genetics ,Other Biological Sciences ,Chemical Physics - Abstract
Early detection of prostate cancer (PC) is largely carried out using assessment of prostate-specific antigen (PSA) level; yet it cannot reliably discriminate between benign pathologies and clinically significant forms of PC. To overcome the current limitations of PSA, new urinary and serum biomarkers have been developed in recent years. Although several biomarkers have been explored in various scenarios and patient settings, to date, specific guidelines with a high level of evidence on the use of these markers are lacking. Recent advances in metabolomic, genomics, and proteomics have made new potential biomarkers available. A number of studies focused on the characterization of the specific PC metabolic phenotype using different experimental approaches has been recently reported; yet, to date, research on metabolomic application for PC has focused on a small group of metabolites that have been known to be related to the prostate gland. Exosomes are extracellular vesicles that are secreted from all mammalian cells and virtually detected in all bio-fluids, thus allowing their use as tumor biomarkers. Thanks to a general improvement of the technical equipment to analyze exosomes, we are able to obtain reliable quantitative and qualitative information useful for clinical application. Although some pilot clinical investigations have proposed potential PC biomarkers, data are still preliminary and non-conclusive.
- Published
- 2021
14. Randomized Trial of a Vaccine Regimen to Prevent Chronic HCV Infection
- Author
-
Page, Kimberly, Melia, Michael T, Veenhuis, Rebecca T, Winter, Matthew, Rousseau, Kimberly E, Massaccesi, Guido, Osburn, William O, Forman, Michael, Thomas, Elaine, Thornton, Karla, Wagner, Katherine, Vassilev, Ventzislav, Lin, Lan, Lum, Paula J, Giudice, Linda C, Stein, Ellen, Asher, Alice, Chang, Soju, Gorman, Richard, Ghany, Marc G, Liang, T Jake, Wierzbicki, Michael R, Scarselli, Elisa, Nicosia, Alfredo, Folgori, Antonella, Capone, Stefania, and Cox, Andrea L
- Subjects
Vaccine Related ,Liver Disease ,Clinical Trials and Supportive Activities ,Clinical Research ,Hepatitis ,Emerging Infectious Diseases ,Digestive Diseases ,HIV/AIDS ,Hepatitis - C ,Immunization ,Chronic Liver Disease and Cirrhosis ,Infectious Diseases ,Prevention ,Biotechnology ,Prevention of disease and conditions ,and promotion of well-being ,6.1 Pharmaceuticals ,3.4 Vaccines ,Evaluation of treatments and therapeutic interventions ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Infection ,Good Health and Well Being ,Adenoviruses ,Simian ,Adolescent ,Adult ,Animals ,Double-Blind Method ,Female ,Genetic Vectors ,Hepatitis C Antibodies ,Hepatitis C ,Chronic ,Humans ,Immunogenicity ,Vaccine ,Incidence ,Male ,Middle Aged ,Pan troglodytes ,Substance Abuse ,Intravenous ,T-Lymphocytes ,Vaccines ,Synthetic ,Viral Hepatitis Vaccines ,Young Adult ,Medical and Health Sciences ,General & Internal Medicine - Abstract
BackgroundA safe and effective vaccine to prevent chronic hepatitis C virus (HCV) infection is a critical component of efforts to eliminate the disease.MethodsIn this phase 1-2 randomized, double-blind, placebo-controlled trial, we evaluated a recombinant chimpanzee adenovirus 3 vector priming vaccination followed by a recombinant modified vaccinia Ankara boost; both vaccines encode HCV nonstructural proteins. Adults who were considered to be at risk for HCV infection on the basis of a history of recent injection drug use were randomly assigned (in a 1:1 ratio) to receive vaccine or placebo on days 0 and 56. Vaccine-related serious adverse events, severe local or systemic adverse events, and laboratory adverse events were the primary safety end points. The primary efficacy end point was chronic HCV infection, defined as persistent viremia for 6 months.ResultsA total of 548 participants underwent randomization, with 274 assigned to each group. There was no significant difference in the incidence of chronic HCV infection between the groups. In the per-protocol population, chronic HCV infection developed in 14 participants in each group (hazard ratio [vaccine vs. placebo], 1.53; 95% confidence interval [CI], 0.66 to 3.55; vaccine efficacy, -53%; 95% CI, -255 to 34). In the modified intention-to-treat population, chronic HCV infection developed in 19 participants in the vaccine group and 17 in placebo group (hazard ratio, 1.66; 95% CI, 0.79 to 3.50; vaccine efficacy, -66%; 95% CI, -250 to 21). The geometric mean peak HCV RNA level after infection differed between the vaccine group and the placebo group (152.51×103 IU per milliliter and 1804.93×103 IU per milliliter, respectively). T-cell responses to HCV were detected in 78% of the participants in the vaccine group. The percentages of participants with serious adverse events were similar in the two groups.ConclusionsIn this trial, the HCV vaccine regimen did not cause serious adverse events, produced HCV-specific T-cell responses, and lowered the peak HCV RNA level, but it did not prevent chronic HCV infection. (Funded by the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov number, NCT01436357.).
- Published
- 2021
15. Parallel studies of mucosal immunity in the reproductive and gastrointestinal mucosa of HIV‐infected women
- Author
-
Shanmugasundaram, Uma, Critchfield, J William, Giudice, Linda C, Smith‐McCune, Karen, Greenblatt, Ruth M, and Shacklett, Barbara L
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Immunology ,Clinical Research ,Digestive Diseases ,Infectious Diseases ,HIV/AIDS ,Contraception/Reproduction ,Infection ,Good Health and Well Being ,Adult ,Antiretroviral Therapy ,Highly Active ,CD4-Positive T-Lymphocytes ,Cells ,Cultured ,Female ,Genitalia ,Female ,HIV Infections ,HIV-1 ,Humans ,Immunity ,Mucosal ,Immunophenotyping ,Intestinal Mucosa ,Lymphocyte Activation ,Middle Aged ,Organ Specificity ,CTL ,endocervix ,endometrium ,GALT ,HIV ,MALT ,T-cell - Abstract
ProblemThe effects of HIV on the gastrointestinal tract (GIT), including CD4 depletion, epithelial disruption, and collagen deposition, are well documented and only partially reversed by combination antiretroviral therapy (cART). However, the effects of HIV on the female reproductive tract (FRT) are poorly understood, and most studies have focused on ectocervix and vagina without assessing the upper tract. Here, we investigated CD4+ T-cell frequency, phenotype, and HIV-specific T-cell responses in the endocervix and endometrium of HIV-infected women, comparing these tissues to the GIT.Method of studyMucosal samples and blood were obtained from 18 women: four who were HIV-positive and not on cART for at least 3 years prior to sampling, including two natural controllers (viral load [VL] undetectable and CD4 >350); nine women on cART with low to undetectable VL; and five HIV-uninfected women. Mucosal samples included terminal ileum, sigmoid colon, endocervical cytobrush, endocervical curettage, and endometrial biopsy. T-cell frequency, phenotypes, and HIV-specific T-cell responses were analyzed by multiparameter flow cytometry.ResultsT-cell activation, measured by CD38/HLA-DR co-expression, remained significantly elevated in endometrium following cART, but was lower in gastrointestinal tissues. HIV-specific CD8+ T-cell responses were detected in ileum, colon, and endometrial tissues of women both on and off cART, and were of higher magnitude on those not on cART.ConclusionOur findings reveal differences in CD4+ T-cell frequencies, immune activation, and HIV-specific T-cell responses between the gastrointestinal and reproductive tracts, and highlight differences between HIV controllers and women on cART.
- Published
- 2020
16. Steroid hormones regulate genome-wide epigenetic programming and gene transcription in human endometrial cells with marked aberrancies in endometriosis.
- Author
-
Houshdaran, Sahar, Oke, Ashwini B, Fung, Jennifer C, Vo, Kim Chi, Nezhat, Camran, and Giudice, Linda C
- Subjects
Endometrium ,Chromatin ,Humans ,Endometriosis ,Estradiol ,Progesterone ,DNA Methylation ,Epigenesis ,Genetic ,CpG Islands ,Adult ,Female ,Transcriptome ,Epigenesis ,Genetic ,Genetics ,Developmental Biology - Abstract
Programmed cellular responses to cycling ovarian-derived steroid hormones are central to normal endometrial function. Abnormalities therein, as in the estrogen-dependent, progesterone-"resistant" disorder, endometriosis, predispose to infertility and poor pregnancy outcomes. The endometrial stromal fibroblast (eSF) is a master regulator of pregnancy success. However, the complex hormone-epigenome-transcriptome interplay in eSF by each individual steroid hormone, estradiol (E2) and/or progesterone (P4), under physiologic and pathophysiologic conditions, is poorly understood and was investigated herein. Genome-wide analysis in normal, early and late stage eutopic eSF revealed: i) In contrast to P4, E2 extensively affected the eSF DNA methylome and transcriptome. Importantly, E2 resulted in a more open versus closed chromatin, confirmed by histone modification analysis. Combined E2 with P4 affected a totally different landscape than E2 or P4 alone. ii) P4 responses were aberrant in early and late stage endometriosis, and mapping differentially methylated CpG sites with progesterone receptor targets from the literature revealed different but not decreased P4-targets, leading to question the P4-"resistant" phenotype in endometriosis. Interestingly, an aberrant E2-response was noted in eSF from endometriosis women; iii) Steroid hormones affected specific genomic contexts and locations, significantly enriching enhancers and intergenic regions and minimally involving proximal promoters and CpG islands, regardless of hormone type and eSF disease state. iv) In eSF from women with endometriosis, aberrant hormone-induced methylation signatures were mainly due to existing DNA methylation marks prior to hormone treatments and involved known endometriosis genes and pathways. v) Distinct DNA methylation and transcriptomic signatures revealed early and late stage endometriosis comprise unique disease subtypes. Taken together, the data herein, for the first time, provide significant insight into the hormone-epigenome-transcriptome interplay of each steroid hormone in normal eSF, and aberrant E2 response, distinct disease subtypes, and pre-existing epigenetic aberrancies in the setting of endometriosis, provide mechanistic insights into how endometriosis affects endometrial function/dysfunction.
- Published
- 2020
17. Adenomyosis: Mechanisms and Pathogenesis
- Author
-
Zhai, Junyu, Vannuccini, Silvia, Petraglia, Felice, and Giudice, Linda C
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Stem Cell Research ,Cancer ,Uterine Cancer ,Pain Research ,Infertility ,Contraception/Reproduction ,Genetics ,Endometriosis ,2.1 Biological and endogenous factors ,Aetiology ,Reproductive health and childbirth ,Adenomyosis ,Animals ,Cell Movement ,Dysmenorrhea ,Endometrium ,Female ,Humans ,Infertility ,Female ,Menorrhagia ,Myometrium ,adenomyosis ,mechanisms ,pathogenesis ,metaplasia ,endometrial-myometrial interface ,Paediatrics and Reproductive Medicine ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Abstract
Adenomyosis is a common disorder of the uterus, and is associated with an enlarged uterus, heavy menstrual bleeding (HMB), pelvic pain, and infertility. It is characterized by endometrial epithelial cells and stromal fibroblasts abnormally found in the myometrium where they elicit hyperplasia and hypertrophy of surrounding smooth muscle cells. While both the mechanistic processes and the pathogenesis of adenomyosis are uncertain, several theories have been put forward addressing how this disease develops. These include intrinsic or induced (1) microtrauma of the endometrial-myometrial interface; (2) enhanced invasion of endometrium into myometrium; (3) metaplasia of stem cells in myometrium; (4) infiltration of endometrial cells in retrograde menstrual effluent into the uterine wall from the serosal side; (5) induction of adenomyotic lesions by aberrant local steroid and pituitary hormones; and (6) abnormal uterine development in response to genetic and epigenetic modifications. Dysmenorrhea, HMB, and infertility are likely results of inflammation, neurogenesis, angiogenesis, and contractile abnormalities in the endometrial and myometrial components. Elucidating mechanisms underlying the pathogenesis of adenomyosis raise possibilities to develop targeted therapies to ameliorate symptoms beyond the current agents that are largely ineffective. Herein, we address these possible etiologies and data that support underlying mechanisms.
- Published
- 2020
18. Progestins Related to Progesterone and Testosterone Elicit Divergent Human Endometrial Transcriptomes and Biofunctions.
- Author
-
Houshdaran, Sahar, Chen, Joseph C, Vallvé-Juanico, Júlia, Balayan, Shayna, Vo, Kim Chi, Smith-McCune, Karen, Greenblatt, Ruth M, Irwin, Juan C, and Giudice, Linda C
- Subjects
Endometrium ,Fibroblasts ,Humans ,Testosterone ,Progesterone ,Estrogens ,Progestins ,Cell Survival ,Gene Expression Regulation ,Female ,angiogenesis ,endometrial stromal fibroblasts ,inflammation ,progestins ,transcriptome ,Other Chemical Sciences ,Genetics ,Other Biological Sciences ,Chemical Physics - Abstract
Progestins are widely used for the treatment of gynecologic disorders and alone, or combined with an estrogen, are used as contraceptives. While their potencies, efficacies and side effects vary due to differences in structures, doses and routes of administration, little is known about their effects on the endometrial transcriptome in the presence or absence of estrogen. Herein, we assessed the transcriptome and pathways induced by progesterone (P4) and the three most commonly used synthetic progestins, medroxyprogesterone acetate (MPA), levonorgestrel (LNG), and norethindrone acetate (NETA), on human endometrial stromal fibroblasts (eSF), key players in endometrial physiology and reproductive success. While there were similar transcriptional responses, each progestin induced unique genes and biofunctions, consistent with their structural similarities to progesterone (P4 and MPA) or testosterone (LNG and NETA), involving cellular proliferation, migration and invasion. Addition of estradiol (E2) to each progestin influenced the number of differentially expressed genes and biofunctions in P4 and MPA, while LNG and NETA signatures were more independent of E2. Together, these data suggest different mechanisms of action for different progestins, with progestin-specific altered signatures when combined with E2. Further investigation is warranted for a personalized approach in different gynecologic disorders, for contraception, and minimizing side effects associated with their use.
- Published
- 2020
19. Seminal plasma promotes decidualization of endometrial stromal fibroblasts in vitro from women with and without inflammatory disorders in a manner dependent on interleukin-11 signaling.
- Author
-
George, Ashley F, Jang, Karen S, Nyegaard, Mette, Neidleman, Jason, Spitzer, Trimble L, Xie, Guorui, Chen, Joseph C, Herzig, Eytan, Laustsen, Anders, Marques de Menezes, Erika G, Houshdaran, Sahar, Pilcher, Christopher D, Norris, Philip J, Jakobsen, Martin R, Greene, Warner C, Giudice, Linda C, and Roan, Nadia R
- Subjects
Contraception/Reproduction ,Clinical Research ,2.1 Biological and endogenous factors ,Aetiology ,Reproductive health and childbirth ,Cross-Sectional Studies ,Decidua ,Endometriosis ,Endometrium ,Female ,Fibroblasts ,Humans ,Interleukin-11 ,Polycystic Ovary Syndrome ,Semen ,reproduction ,semen ,endometrium ,stromal fibroblast ,decidualization ,RNAseq ,interleukin-II ,extracellular vesicles ,CRISPR ,Cas-9 ,CRISPR/Cas-9 ,interleukin-11 ,Medical and Health Sciences ,Studies in Human Society ,Obstetrics & Reproductive Medicine - Abstract
Study questionDo seminal plasma (SP) and its constituents affect the decidualization capacity and transcriptome of human primary endometrial stromal fibroblasts (eSFs)?Summary answerSP promotes decidualization of eSFs from women with and without inflammatory disorders (polycystic ovary syndrome (PCOS), endometriosis) in a manner that is not mediated through semen amyloids and that is associated with a potent transcriptional response, including the induction of interleukin (IL)-11, a cytokine important for SP-induced decidualization.What is known alreadyClinical studies have suggested that SP can promote implantation, and studies in vitro have demonstrated that SP can promote decidualization, a steroid hormone-driven program of eSF differentiation that is essential for embryo implantation and that is compromised in women with the inflammatory disorders PCOS and endometriosis.Study design, size, durationThis is a cross-sectional study involving samples treated with vehicle alone versus treatment with SP or SP constituents. SP was tested for the ability to promote decidualization in vitro in eSFs from women with or without PCOS or endometriosis (n = 9). The role of semen amyloids and fractionated SP in mediating this effect and in eliciting transcriptional changes in eSFs was then studied. Finally, the role of IL-11, a cytokine with a key role in implantation and decidualization, was assessed as a mediator of the SP-facilitated decidualization.Participants/materials, setting, methodseSFs and endometrial epithelial cells (eECs) were isolated from endometrial biopsies from women of reproductive age undergoing benign gynecologic procedures and maintained in vitro. Assays were conducted to assess whether the treatment of eSFs with SP or SP constituents affects the rate and extent of decidualization in women with and without inflammatory disorders. To characterize the response of the endometrium to SP and SP constituents, RNA was isolated from treated eSFs or eECs and analyzed by RNA sequencing (RNAseq). Secreted factors in conditioned media from treated cells were analyzed by Luminex and ELISA. The role of IL-11 in SP-induced decidualization was assessed through Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas-9-mediated knockout experiments in primary eSFs.Main results and the role of chanceSP promoted decidualization both in the absence and presence of steroid hormones (P
- Published
- 2020
20. HIV efficiently infects T cells from the endometrium and remodels them to promote systemic viral spread
- Author
-
Ma, Tongcui, Luo, Xiaoyu, George, Ashley F, Mukherjee, Gourab, Sen, Nandini, Spitzer, Trimble L, Giudice, Linda C, Greene, Warner C, and Roan, Nadia R
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Immunology ,Prevention ,HIV/AIDS ,Sexually Transmitted Infections ,Women's Health ,Infectious Diseases ,2.2 Factors relating to the physical environment ,2.1 Biological and endogenous factors ,Aetiology ,Infection ,Good Health and Well Being ,Adult ,CD4-Positive T-Lymphocytes ,Endometrium ,Female ,HIV ,HIV Infections ,Host-Pathogen Interactions ,Humans ,Middle Aged ,Survivin ,Young Adult ,female reproductive tract ,human ,infectious disease ,microbiology ,mucosa ,t cells ,virus ,viruses ,Biochemistry and Cell Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
The female reproductive tract (FRT) is the most common site of infection during HIV transmission to women, but viral remodeling complicates characterization of cells targeted for infection. Here, we report extensive phenotypic analyses of HIV-infected endometrial cells by CyTOF, and use a 'nearest neighbor' bioinformatics approach to trace cells to their original pre-infection phenotypes. Like in blood, HIV preferentially targets memory CD4+ T cells in the endometrium, but these cells exhibit unique phenotypes and sustain much higher levels of infection. Genital cell remodeling by HIV includes downregulating TCR complex components and modulating chemokine receptor expression to promote dissemination of infected cells to lymphoid follicles. HIV also upregulates the anti-apoptotic protein BIRC5, which when blocked promotes death of infected endometrial cells. These results suggest that HIV remodels genital T cells to prolong viability and promote viral dissemination and that interfering with these processes might reduce the likelihood of systemic viral spread.
- Published
- 2020
21. Macrophages display proinflammatory phenotypes in the eutopic endometrium of women with endometriosis with relevance to an infectious etiology of the disease
- Author
-
Vallvé-Juanico, Júlia, Santamaria, Xavier, Vo, Kim Chi, Houshdaran, Sahar, and Giudice, Linda C
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Clinical Research ,Contraception/Reproduction ,Genetics ,Endometriosis ,Pain Research ,Aetiology ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Underpinning research ,Reproductive health and childbirth ,Good Health and Well Being ,Adult ,Case-Control Studies ,Cell Plasticity ,Cell Separation ,Cellular Microenvironment ,Endometrium ,Female ,Flow Cytometry ,Gene Expression Profiling ,Gene Expression Regulation ,Humans ,Macrophage Activation ,Macrophages ,Middle Aged ,Phenotype ,Prospective Studies ,RNA-Seq ,Signal Transduction ,Transcriptome ,Young Adult ,macrophages ,inflammation ,endometrium ,infection ,Clinical Sciences ,Paediatrics and Reproductive Medicine ,Public Health and Health Services ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Abstract
ObjectiveTo phenotype transcriptomically M1 macrophages (Mϕ1) and M2 macrophages (Mϕ2) in the endometrium of women with endometriosis.DesignProspective experimental study.SettingUniversity research laboratory.Patient(s)Six women with endometriosis and five controls without disease, in the secretory phase of the menstrual cycle.Intervention(s)Mϕ1, Mϕ2, uterine natural killer, and T regulatory cells were isolated from human endometrium using a uniquely designed cell-specific fluorescence activating cell sorting panel. Transcriptome profiles were assessed by RNA high sequencing, bioinformatics, and biological pathway analyses.Main outcomes measure(s)Differential gene expression between Mϕ1 and Mϕ2 in women with and without endometriosis and in Mϕ1 versus Mϕ2 in each group was determined and involved different biologic and signaling pathways.Result(s)Flow cytometry analysis showed no significant differences in total numbers of leukocytes between control and endometriosis groups, although Mϕ1 were higher in the endometriosis group versus controls. Statistical transcriptomic analysis was performed only in Mϕ1 and Mϕ2 populations due to larger sample sizes. Bioinformatic analyses revealed that in women with endometriosis, endometrial Mϕ1 are more proinflammatory than controls and that Mϕ2 paradoxically have a proinflammatory phenotype.Conclusion(s)As Mϕ are phenotypically plastic and their polarization state depends on their microenvironment, the altered endometrial environment in women with endometriosis may promote endometrial Mϕ2 polarization and an Mϕ1 proinflammatory phenotype. Moreover, aberrant phenotypes of Mϕ may contribute to abnormal gene expression of the eutopic endometrium and a proinflammatory environment in women with endometriosis relevant to the pathophysiology of the disease and compromised reproductive outcomes.
- Published
- 2019
22. The endometrial immune environment of women with endometriosis
- Author
-
Vallvé-Juanico, Júlia, Houshdaran, Sahar, and Giudice, Linda C
- Subjects
Endometriosis ,Infertility ,Contraception/Reproduction ,Clinical Research ,Pain Research ,1.1 Normal biological development and functioning ,Underpinning research ,2.1 Biological and endogenous factors ,Aetiology ,Reproductive health and childbirth ,Dendritic Cells ,Endometrium ,Estrogens ,Female ,Humans ,Inflammation ,Killer Cells ,Natural ,Macrophages ,Pregnancy ,T-Lymphocytes ,Regulatory ,endometrium ,endometriosis ,immune populations ,immune cell markers ,inflammation ,Clinical Sciences ,Paediatrics and Reproductive Medicine ,Public Health and Health Services ,Obstetrics & Reproductive Medicine - Abstract
BackgroundEndometriosis, a common oestrogen-dependent inflammatory disorder in women of reproductive age, is characterized by endometrial-like tissue outside its normal location in the uterus, which causes pelvic scarring, pain and infertility. While its pathogenesis is poorly understood, the immune system (systemically and locally in endometrium, pelvic endometriotic lesions and peritoneal fluid) is believed to play a central role in its aetiology, pathophysiology and associated morbidities of pain, infertility and poor pregnancy outcomes. However, immune cell populations within the endometrium of women with the disease have had incomplete phenotyping, thereby limiting insight into their roles in this disorder.Objective and rationaleThe objective herein was to determine reproducible and consistent findings regarding specific immune cell populations and their abundance, steroid hormone responsiveness, functionality, activation states, and markers, locally and systemically in women with and without endometriosis.Search methodsA comprehensive English language PubMed, Medline and Google Scholar search was conducted with key search terms that included endometriosis, inflammation, human eutopic/ectopic endometrium, immune cells, immune population, immune system, macrophages, dendritic cells (DC), natural killer cells, mast cells, eosinophils, neutrophils, B cells and T cells.OutcomesIn women with endometriosis compared to those without endometriosis, some endometrial immune cells display similar cycle-phase variation, whereas macrophages (Mø), immature DC and regulatory T cells behave differently. A pro-inflammatory Mø1 phenotype versus anti-inflammatory Mø2 phenotype predominates and natural killer cells display abnormal activity in endometrium of women with the disease. Conflicting data largely derive from small studies, variably defined hormonal milieu and different experimental approaches and technologies.Wider implicationsPhenotyping immune cell subtypes is essential to determine the role of the endometrial immune niche in pregnancy and endometrial homeostasis normally and in women with poor reproductive history and can facilitate development of innovative diagnostics and therapeutics for associated symptoms and compromised reproductive outcomes.
- Published
- 2019
23. MC1R variants in childhood and adolescent melanoma: a retrospective pooled analysis of a multicentre cohort
- Author
-
Pellegrini, Cristina, Botta, Francesca, Massi, Daniela, Martorelli, Claudia, Facchetti, Fabio, Gandini, Sara, Maisonneuve, Patrick, Avril, Marie-Françoise, Demenais, Florence, Paillerets, Brigitte Bressac-de, Hoiom, Veronica, Cust, Anne E, Anton-Culver, Hoda, Gruber, Stephen B, Gallagher, Richard P, Marrett, Loraine, Zanetti, Roberto, Dwyer, Terence, Thomas, Nancy E, Begg, Colin B, Berwick, Marianne, Puig, Susana, Potrony, Miriam, Nagore, Eduardo, Ghiorzo, Paola, Menin, Chiara, Manganoni, Ausilia Maria, Rodolfo, Monica, Brugnara, Sonia, Passoni, Emanuela, Sekulovic, Lidija Kandolf, Baldini, Federica, Guida, Gabriella, Stratigos, Alexandros, Ozdemir, Fezal, Ayala, Fabrizio, Fernandez-de-Misa, Ricardo, Quaglino, Pietro, Ribas, Gloria, Romanini, Antonella, Migliano, Emilia, Stanganelli, Ignazio, Kanetsky, Peter A, Pizzichetta, Maria Antonietta, García-Borrón, Jose Carlos, Nan, Hongmei, Landi, Maria Teresa, Little, Julian, Newton-Bishop, Julia, Sera, Francesco, Fargnoli, Maria Concetta, Raimondi, Sara, Group, IMI Study, Alaibac, Mauro, Ferrari, Andrea, Valeri, Barbara, Sicher, Mariacristina, Mangiola, Daniela, Nazzaro, Gianluca, Tosti, Giulio, Mazzarol, Giovanni, Giudice, Giuseppe, Ribero, Simone, Astrua, Chiara, Mazzoni, Laura, Group, GEM Study, Orlow, Irene, Mujumdar, Urvi, Hummer, Amanda, Busam, Klaus, Roy, Pampa, Canchola, Rebecca, Clas, Brian, Cotignola, Javiar, Monroe, Yvette, Armstrong, Bruce, Kricker, Anne, Litchfield, Melisa, Tucker, Paul, Stephens, Nicola, Switzer, Teresa, Theis, Beth, From, Lynn, Chowdhury, Noori, Vanasse, Louise, Purdue, Mark, Northrup, David, Rosso, Stefano, Sacerdote, Carlotta, Leighton, Nancy, Gildea, Maureen, Bonner, Joe, Jeter, Joanne, Klotz, Judith, Wilcox, Homer, Weiss, Helen, Millikan, Robert, Mattingly, Dianne, and Player, Jon
- Subjects
Clinical Research ,Pediatric ,Genetics ,Prevention ,Cancer ,Adolescent ,Adult ,Aged ,Biomarkers ,Tumor ,Case-Control Studies ,Child ,Female ,Genetic Predisposition to Disease ,Germ-Line Mutation ,Humans ,Logistic Models ,Male ,Melanoma ,Middle Aged ,Odds Ratio ,Polymorphism ,Genetic ,Receptor ,Melanocortin ,Type 1 ,Retrospective Studies ,Skin Neoplasms ,IMI Study Group ,GEM Study Group ,M-SKIP Study Group - Abstract
BackgroundGermline variants in the melanocortin 1 receptor gene (MC1R) might increase the risk of childhood and adolescent melanoma, but a clear conclusion is challenging because of the low number of studies and cases. We assessed the association of MC1R variants with childhood and adolescent melanoma in a large study comparing the prevalence of MC1R variants in child or adolescent patients with melanoma to that in adult patients with melanoma and in healthy adult controls.MethodsIn this retrospective pooled analysis, we used the M-SKIP Project, the Italian Melanoma Intergroup, and other European groups (with participants from Australia, Canada, France, Greece, Italy, the Netherlands, Serbia, Spain, Sweden, Turkey, and the USA) to assemble an international multicentre cohort. We gathered phenotypic and genetic data from children or adolescents diagnosed with sporadic single-primary cutaneous melanoma at age 20 years or younger, adult patients with sporadic single-primary cutaneous melanoma diagnosed at age 35 years or older, and healthy adult individuals as controls. We calculated odds ratios (ORs) for childhood and adolescent melanoma associated with MC1R variants by multivariable logistic regression. Subgroup analysis was done for children aged 18 or younger and 14 years or younger.FindingsWe analysed data from 233 young patients, 932 adult patients, and 932 healthy adult controls. Children and adolescents had higher odds of carrying MC1R r variants than did adult patients (OR 1·54, 95% CI 1·02-2·33), including when analysis was restricted to patients aged 18 years or younger (1·80, 1·06-3·07). All investigated variants, except Arg160Trp, tended, to varying degrees, to have higher frequencies in young patients than in adult patients, with significantly higher frequencies found for Val60Leu (OR 1·60, 95% CI 1·05-2·44; p=0·04) and Asp294His (2·15, 1·05-4·40; p=0·04). Compared with those of healthy controls, young patients with melanoma had significantly higher frequencies of any MC1R variants.InterpretationOur pooled analysis of MC1R genetic data of young patients with melanoma showed that MC1R r variants were more prevalent in childhood and adolescent melanoma than in adult melanoma, especially in patients aged 18 years or younger. Our findings support the role of MC1R in childhood and adolescent melanoma susceptibility, with a potential clinical relevance for developing early melanoma detection and preventive strategies.FundingSPD-Pilot/Project-Award-2015; AIRC-MFAG-11831.
- Published
- 2019
24. Mutations in MAST1 Cause Mega-Corpus-Callosum Syndrome with Cerebellar Hypoplasia and Cortical Malformations
- Author
-
Tripathy, Ratna, Leca, Ines, van Dijk, Tessa, Weiss, Janneke, van Bon, Bregje W, Sergaki, Maria Christina, Gstrein, Thomas, Breuss, Martin, Tian, Guoling, Bahi-Buisson, Nadia, Paciorkowski, Alexander R, Pagnamenta, Alistair T, Wenninger-Weinzierl, Andrea, Martinez-Reza, Maria Fernanda, Landler, Lukas, Lise, Stefano, Taylor, Jenny C, Terrone, Gaetano, Vitiello, Giuseppina, Del Giudice, Ennio, Brunetti-Pierri, Nicola, D’Amico, Alessandra, Reymond, Alexandre, Voisin, Norine, Bernstein, Jonathan A, Farrelly, Ellyn, Kini, Usha, Leonard, Thomas A, Valence, Stéphanie, Burglen, Lydie, Armstrong, Linlea, Hiatt, Susan M, Cooper, Gregory M, Aldinger, Kimberly A, Dobyns, William B, Mirzaa, Ghayda, Pierson, Tyler Mark, Baas, Frank, Chelly, Jamel, Cowan, Nicholas J, and Keays, David Anthony
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Rare Diseases ,Pediatric ,Genetics ,Brain Disorders ,Intellectual and Developmental Disabilities (IDD) ,Congenital Structural Anomalies ,2.1 Biological and endogenous factors ,Aetiology ,Neurological ,Agenesis of Corpus Callosum ,Animals ,Animals ,Newborn ,Apoptosis ,Brain ,Cells ,Cultured ,Cerebellum ,Child ,Developmental Disabilities ,Disease Models ,Animal ,Embryo ,Mammalian ,Female ,Gene Expression Regulation ,Developmental ,Humans ,Male ,Malformations of Cortical Development ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,Microtubule-Associated Proteins ,Mutation ,Nerve Tissue Proteins ,Nervous System Malformations ,PAX6 Transcription Factor ,MAST1 ,cerebellar hypoplasia ,corpus callosum ,microdeletion ,microtubules ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery ,Biological psychology - Abstract
Corpus callosum malformations are associated with a broad range of neurodevelopmental diseases. We report that de novo mutations in MAST1 cause mega-corpus-callosum syndrome with cerebellar hypoplasia and cortical malformations (MCC-CH-CM) in the absence of megalencephaly. We show that MAST1 is a microtubule-associated protein that is predominantly expressed in post-mitotic neurons and is present in both dendritic and axonal compartments. We further show that Mast1 null animals are phenotypically normal, whereas the deletion of a single amino acid (L278del) recapitulates the distinct neurological phenotype observed in patients. In animals harboring Mast1 microdeletions, we find that the PI3K/AKT3/mTOR pathway is unperturbed, whereas Mast2 and Mast3 levels are diminished, indicative of a dominant-negative mode of action. Finally, we report that de novo MAST1 substitutions are present in patients with autism and microcephaly, raising the prospect that mutations in this gene give rise to a spectrum of neurodevelopmental diseases.
- Published
- 2018
25. Potent and rapid activation of tropomyosin-receptor kinase A in endometrial stromal fibroblasts by seminal plasma.
- Author
-
Martin, Jeremy W, Chen, Joseph C, Neidleman, Jason, Tatsumi, Keiji, Hu, James, Giudice, Linda C, Greene, Warner C, and Roan, Nadia R
- Subjects
Endometrium ,Fibroblasts ,Stromal Cells ,Semen ,Humans ,Receptor ,trkA ,Signal Transduction ,Phosphorylation ,Embryo Implantation ,Adult ,Female ,endometrium ,seminal plasma ,cytokines ,fertility ,signal transduction ,Receptor ,trkA ,Medical and Health Sciences ,Biological Sciences ,Obstetrics & Reproductive Medicine - Abstract
Seminal plasma (SP), the liquid fraction of semen, is not mandatory for conception, but clinical studies suggest that SP improves implantation rates. Prior in vitro studies examining the effects of SP on the endometrium, the site of implantation, surprisingly revealed that SP induces transcriptional profiles associated with neurogenesis. We investigated the presence and activity of neurogenesis pathways in the endometrium, focusing on TrkA, one of the canonical receptors associated with neurotrophic signaling. We demonstrate that TrkA is expressed in the endometrium. To determine if SP activates TrkA signaling, we isolated the two most abundant endometrial cell types-endometrial epithelial cells (eEC) and endometrial stromal fibroblasts (eSF)-and examined TrkA activity in these cells after SP exposure. While SP only moderately activated TrkA in eEC, it potently and rapidly activated TrkA in eSF. This activation occurred in both non-decidualized and decidualized eSF. Blocking this pathway resulted in dysregulation of SP-induced cytokine production by eSF. Surprisingly, while the canonical TrkA agonist nerve growth factor was detected in SP, TrkA activation was principally induced by a 30-100-kDa protein whose identity remains to be established. Our results show that TrkA signaling is highly active in eSF and is rapidly induced by SP.
- Published
- 2018
26. In vitro evidence that platelet-rich plasma stimulates cellular processes involved in endometrial regeneration
- Author
-
Aghajanova, Lusine, Houshdaran, Sahar, Balayan, Shaina, Manvelyan, Evelina, Irwin, Juan C, Huddleston, Heather G, and Giudice, Linda C
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Regenerative Medicine ,Stem Cell Research ,Uterine Cancer ,Cancer ,Clinical Research ,Stem Cell Research - Nonembryonic - Human ,Stem Cell Research - Nonembryonic - Non-Human ,Development of treatments and therapeutic interventions ,5.2 Cellular and gene therapies ,Aged ,Cell Movement ,Cell Proliferation ,Cells ,Cultured ,Endometrium ,Epithelial-Mesenchymal Transition ,Female ,Fibroblasts ,Gene Expression Regulation ,Humans ,Male ,Mesenchymal Stem Cells ,Platelet-Rich Plasma ,Regeneration ,Platelet-rich plasma ,Stem cells ,Proliferation ,Genetics ,Paediatrics and Reproductive Medicine ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Abstract
PURPOSE:The study aims to test the hypothesis that platelet-rich plasma (PRP) stimulates cellular processes involved in endometrial regeneration relevant to clinical management of poor endometrial growth or intrauterine scarring. METHODS:Human endometrial stromal fibroblasts (eSF), endometrial mesenchymal stem cells (eMSC), bone marrow-derived mesenchymal stem cells (BM-MSC), and Ishikawa endometrial adenocarcinoma cells (IC) were cultured with/without 5% activated (a) PRP, non-activated (na) PRP, aPPP (platelet-poor-plasma), and naPPP. Treatment effects were evaluated with cell proliferation (WST-1), wound healing, and chemotaxis Transwell migration assays. Mesenchymal-to-epithelial transition (MET) was evaluated by cytokeratin and vimentin expression. Differential gene expression of various markers was analyzed by multiplex Q-PCR. RESULTS:Activated PRP enhanced migration of all cell types, compared to naPRP, aPPP, naPPP, and vehicle controls, in a time-dependent manner (p
- Published
- 2018
27. Evaluation, validation and refinement of noninvasive diagnostic biomarkers for endometriosis (ENDOmarker): A protocol to phenotype bio-specimens for discovery and validation
- Author
-
Barnhart, Kurt, Giudice, Linda, Young, Steve, Thomas, Tracey, Diamond, Michael P, Segars, James, Youssef, Wahid A, Krawetz, Stephen, Santoro, Nanette, Eisenberg, Esther, Zhang, Heping, and Network, for the NICHD Cooperative Reproductive Medicine
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Clinical Sciences ,Infertility ,Clinical Research ,Clinical Trials and Supportive Activities ,Contraception/Reproduction ,Patient Safety ,Endometriosis ,Prevention ,Pain Research ,Chronic Pain ,4.1 Discovery and preclinical testing of markers and technologies ,Detection ,screening and diagnosis ,Reproductive health and childbirth ,Good Health and Well Being ,Adult ,Biomarkers ,Conservative Treatment ,Cytokines ,Endometrium ,Female ,Genitalia ,Humans ,Phenotype ,Reproducibility of Results ,Specimen Handling ,Micro RNA ,Phenotyping ,NICHD Cooperative Reproductive Medicine Network ,Medical and Health Sciences ,General Clinical Medicine ,Public Health ,Biomedical and clinical sciences ,Health sciences - Abstract
OBJECTIVE:Endometriosis is a chronic, estrogen dependent condition that affects 5-10% of reproductive aged women and is associated with pelvic pain and infertility. As the approach to therapy shifts from surgical ablation to pharmacological control, a non-surgical mode of diagnosis would be desirable. The ENDOmarker study was designed by the NICHD Reproductive Medicine Network (RMN) to obtain well characterized and phenotyped bio specimens in a standardized fashion from women with and without endometriosis. DESIGN:Development of a diagnostic test. SETTING:Academic medical centers. PATIENTS:This study will enroll up to 500 participants, and follow them for up to 5 months. Included subjects are aged 18-44, scheduled to undergo gynecologic surgery (laparoscopy/laparotomy) for clinical reasons. INTERVENTIONS:Presence and stage of endometriosis (or its absence) is characterized by visual examination at the time of surgery. Subjects will undergo extensive clinical evaluation pre-operatively and at visits one and four months postoperatively. Endometrial biopsy, blood, urine and disease specific questionnaires will be collected at each visit. MAIN OUTCOME:Samples will be placed in a bio-repository to be used to validate and optimize the clinical use of genomic classifiers of the endometrium alone or in combination with serum cytokines as a non-surgical composite marker of endometriosis. CONCLUSION:This protocol can serve as a reference for objective collection of high quality bio specimens for discovery or validation of potential nonsurgical diagnosis of presence or severity of disease.
- Published
- 2018
28. Individual differences in developmental plasticity: A role for early androgens?
- Author
-
Del Giudice, Marco, Barrett, Emily S, Belsky, Jay, Hartman, Sarah, Martel, Michelle M, Sangenstedt, Susanne, and Kuzawa, Christopher W
- Subjects
Animals ,Humans ,Prenatal Exposure Delayed Effects ,Androgens ,Sexual Behavior ,Individuality ,Sex Factors ,Adaptation ,Physiological ,Pregnancy ,Phenotype ,Female ,Male ,Biological Evolution ,Developmental plasticity ,Differential susceptibility ,Sex differences ,Sexual selection ,Testosterone ,Behavioral and Social Science ,Pediatric Research Initiative ,Pediatric ,Aetiology ,2.3 Psychological ,social and economic factors ,Generic health relevance ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Psychiatry - Abstract
Developmental plasticity is a widespread property of living organisms, but different individuals in the same species can vary greatly in how susceptible they are to environmental influences. In humans, research has sought to link variation in plasticity to physiological traits such as stress reactivity, exposure to prenatal stress-related hormones such as cortisol, and specific genes involved in major neurobiological pathways. However, the determinants of individual differences in plasticity are still poorly understood. Here we present the novel hypothesis that, in both sexes, higher exposure to androgens during prenatal and early postnatal life should lead to increased plasticity in traits that display greater male variability (i.e., a majority of physical and behavioral traits). First, we review evidence of greater phenotypic variation and higher susceptibility to environmental factors in males; we then consider evolutionary models that explain greater male variability and plasticity as a result of sexual selection. These empirical and theoretical strands converge on the hypothesis that androgens may promote developmental plasticity, at least for traits that show greater male variability. We discuss a number of potential mechanisms that may mediate this effect (including upregulation of neural plasticity), and address the question of whether androgen-induced plasticity is likely to be adaptive or maladaptive. We conclude by offering suggestions for future studies in this area, and considering some research designs that could be used to empirically test our hypothesis.
- Published
- 2018
29. Stromal fibroblasts from perimenopausal endometrium exhibit a different transcriptome than those from the premenopausal endometrium
- Author
-
Erikson, David W, Barragan, Fatima, Piltonen, Terhi T, Chen, Joseph C, Balayan, Shaina, Irwin, Juan C, and Giudice, Linda C
- Subjects
Stem Cell Research ,Clinical Research ,Estrogen ,Aging ,Stem Cell Research - Nonembryonic - Human ,Regenerative Medicine ,Contraception/Reproduction ,Biotechnology ,2.1 Biological and endogenous factors ,Aetiology ,1.1 Normal biological development and functioning ,Underpinning research ,Adult ,Cell Lineage ,Cluster Analysis ,DNA ,Endometrium ,Female ,Fibroblasts ,Gene Expression Regulation ,Humans ,Mesenchymal Stem Cells ,Microarray Analysis ,Middle Aged ,Perimenopause ,Premenopause ,Principal Component Analysis ,RNA ,Transcriptome ,Young Adult ,menopause ,small nucleolar RNA ,microarray ,endometrial mesenchymal stem cell ,fibroblast ,endometrium ,Biological Sciences ,Medical and Health Sciences ,Obstetrics & Reproductive Medicine - Abstract
Human endometrium undergoes extensive regeneration on a cyclic basis in premenopausal women and likely occurs through the contribution of stem/progenitor cells. Menopause results in the permanent cessation of menstrual cycles and is preceded by perimenopause, a period of several years in which endocrine and biological changes occur and is a period of risk for endometrial proliferative disorders. The objectives of this study were to identify endometrial mesenchymal stem cells (eMSC) and endometrial stromal fibroblasts (eSF) in endometrium of perimenopausal women and perform expression profile analysis of perimenopausal eMSC and eSF to gain insight into the biology of stem/progenitor and lineage cell populations during the transition to menopause. Endometrial tissue was collected from perimenopausal and premenopausal women (n = 9 each). Microarray analysis was performed on fluorescence-activated cell sorting-isolated eSF and eMSC, and data were validated by quantitative real-time PCR. Principal component analysis showed that cells clustered into three distinct groups in 3-dimensional space: perimenopausal eMSC and premenopausal eMSC clustered together, while perimenopausal eSF and premenopausal eSF formed two discrete clusters separate from eMSC. Hierarchical clustering revealed a branching pattern consistent with principle clustering analysis results, indicating that eMSC from premenopausal and perimenopausal women exhibit similar transcriptomic signatures. Pathway analysis revealed dysregulation of cytoskeleton, proliferation, and survival pathways in perimenopausal vs. premenopausal eSF. These data demonstrate that cell populations have altered gene expression in perimenopausal vs. premenopausal endometrium, and that perimenopausal eSF had altered pathway activation when compared to premenopausal eSF. This study provides insight into aging endometrium with relevance to function in reproductively older women.
- Published
- 2017
30. Effects of depot-medroxyprogesterone acetate on the immune microenvironment of the human cervix and endometrium: implications for HIV susceptibility
- Author
-
Smith-McCune, KK, Hilton, JF, Shanmugasundaram, U, Critchfield, JW, Greenblatt, RM, Seidman, D, Averbach, S, Giudice, LC, and Shacklett, BL
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Immunology ,Minority Health ,HIV/AIDS ,Sexually Transmitted Infections ,Contraception/Reproduction ,Prevention ,Clinical Research ,Infectious Diseases ,Women's Health ,Infection ,Good Health and Well Being ,Adult ,CD4-Positive T-Lymphocytes ,Cellular Microenvironment ,Cervix Uteri ,Chemokine CCL2 ,Contraceptive Agents ,Delayed-Action Preparations ,Disease Susceptibility ,Endometrium ,Female ,HIV Infections ,Humans ,Interferon-alpha ,Interleukin-1beta ,Interleukin-6 ,Medroxyprogesterone Acetate ,Receptors ,CCR5 ,Young Adult ,Biological Sciences ,Medical and Health Sciences - Abstract
Depot-medroxyprogesterone acetate is a commonly used injectable contraceptive that has been associated with an increased risk of HIV acquisition. This study compares effects of depot-medroxyprogesterone acetate on immune parameters from several upper reproductive tract compartments relevant to HIV-1 susceptibility in repetitive samples from 15 depot-medroxyprogesterone acetate users and 27 women not on hormonal contraceptives. Compared with samples from unexposed women in the mid-luteal phase, depot-medroxyprogesterone acetate use was associated with: increased endocervical concentrations of MCP1 and IFNalpha2; decreased endocervical concentrations of IL1beta and IL6; increased proportions of endometrial CD4+ and CD8+ cells expressing the activation marker HLADR; increased density of endometrial macrophages; and decreased density of endometrial regulatory T cells. Unlike previous reports with samples from the vagina, we did not observe increased expression of the HIV co-receptor CCR5 on CD4+ T cells in the endocervix or endometrium. Our results indicate important differences in anatomic compartments regarding mechanisms by which depot-medroxyprogesterone acetate could be associated with increased risk of HIV acquisition, including increased recruitment of macrophages to the endometrium, decreased levels of pro-inflammatory cytokines in the endocervix possibly leading to enhanced susceptibility to viral infection, and activation of endometrial T cells.
- Published
- 2017
31. Meta-signature of human endometrial receptivity: a meta-analysis and validation study of transcriptomic biomarkers.
- Author
-
Altmäe, Signe, Koel, Mariann, Võsa, Urmo, Adler, Priit, Suhorutšenko, Marina, Laisk-Podar, Triin, Kukushkina, Viktorija, Saare, Merli, Velthut-Meikas, Agne, Krjutškov, Kaarel, Aghajanova, Lusine, Lalitkumar, Parameswaran G, Gemzell-Danielsson, Kristina, Giudice, Linda, Simón, Carlos, and Salumets, Andres
- Subjects
Endometrium ,Humans ,MicroRNAs ,RNA ,Messenger ,Gene Expression Profiling ,Sequence Analysis ,RNA ,Computational Biology ,Embryo Implantation ,Fertility ,Menstrual Cycle ,Adult ,Female ,Gene Regulatory Networks ,Immunity ,Innate ,Exosomes ,Molecular Sequence Annotation ,Transcriptome ,Gene Ontology ,Biomarkers ,RNA ,Messenger ,Sequence Analysis ,Immunity ,Innate ,Biotechnology ,Genetics ,Human Genome ,Uterine Cancer ,Contraception/Reproduction ,Cancer ,Infertility ,4.1 Discovery and preclinical testing of markers and technologies ,Biochemistry and Cell Biology ,Other Physical Sciences - Abstract
Previous transcriptome studies of the human endometrium have revealed hundreds of simultaneously up- and down-regulated genes that are involved in endometrial receptivity. However, the overlap between the studies is relatively small, and we are still searching for potential diagnostic biomarkers. Here we perform a meta-analysis of endometrial-receptivity associated genes on 164 endometrial samples (76 from 'pre-receptive' and 88 from mid-secretory, 'receptive' phase endometria) using a robust rank aggregation (RRA) method, followed by enrichment analysis, and regulatory microRNA prediction. We identify a meta-signature of endometrial receptivity involving 57 mRNA genes as putative receptivity markers, where 39 of these we confirm experimentally using RNA-sequencing method in two separate datasets. The meta-signature genes highlight the importance of immune responses, the complement cascade pathway and the involvement of exosomes in mid-secretory endometrial functions. Bioinformatic prediction identifies 348 microRNAs that could regulate 30 endometrial-receptivity associated genes, and we confirm experimentally the decreased expression of 19 microRNAs with 11 corresponding up-regulated meta-signature genes in our validation experiments. The 57 identified meta-signature genes and involved pathways, together with their regulatory microRNAs could serve as promising and sought-after biomarkers of endometrial receptivity, fertility and infertility.
- Published
- 2017
32. Treatment of Endometriosis-Associated Pain with Elagolix, an Oral GnRH Antagonist
- Author
-
Taylor, Hugh S, Giudice, Linda C, Lessey, Bruce A, Abrao, Mauricio S, Kotarski, Jan, Archer, David F, Diamond, Michael P, Surrey, Eric, Johnson, Neil P, Watts, Nelson B, Gallagher, J Chris, Simon, James A, Carr, Bruce R, Dmowski, W Paul, Leyland, Nicholas, Rowan, Jean P, Duan, W Rachel, Ng, Juki, Schwefel, Brittany, Thomas, James W, Jain, Rita I, and Chwalisz, Kristof
- Subjects
Complementary and Integrative Health ,Contraception/Reproduction ,Chronic Pain ,Clinical Trials and Supportive Activities ,Endometriosis ,Pain Research ,Clinical Research ,Evaluation of treatments and therapeutic interventions ,6.1 Pharmaceuticals ,Reproductive health and childbirth ,Adolescent ,Adult ,Bone Density ,Dose-Response Relationship ,Drug ,Double-Blind Method ,Dysmenorrhea ,Estrogen Antagonists ,Female ,Gonadotropin-Releasing Hormone ,Hot Flashes ,Humans ,Hydrocarbons ,Fluorinated ,Lipids ,Middle Aged ,Pelvic Pain ,Premenopause ,Pyrimidines ,Young Adult ,Medical and Health Sciences ,General & Internal Medicine - Abstract
Endometriosis is a chronic, estrogen-dependent condition that causes dysmenorrhea and pelvic pain. Elagolix, an oral, nonpeptide, gonadotropin-releasing hormone (GnRH) antagonist, produced partial to nearly full estrogen suppression in previous studies.We performed two similar, double-blind, randomized, 6-month phase 3 trials (Elaris Endometriosis I and II [EM-I and EM-II]) to evaluate the effects of two doses of elagolix - 150 mg once daily (lower-dose group) and 200 mg twice daily (higher-dose group) - as compared with placebo in women with surgically diagnosed endometriosis and moderate or severe endometriosis-associated pain. The two primary efficacy end points were the proportion of women who had a clinical response with respect to dysmenorrhea and the proportion who had a clinical response with respect to nonmenstrual pelvic pain at 3 months. Each of these end points was measured as a clinically meaningful reduction in the pain score and a decreased or stable use of rescue analgesic agents, as recorded in a daily electronic diary.A total of 872 women underwent randomization in Elaris EM-I and 817 in Elaris EM-II; of these women, 653 (74.9%) and 632 (77.4%), respectively, completed the intervention. At 3 months, a significantly greater proportion of women who received each elagolix dose met the clinical response criteria for the two primary end points than did those who received placebo. In Elaris EM-I, the percentage of women who had a clinical response with respect to dysmenorrhea was 46.4% in the lower-dose elagolix group and 75.8% in the higher-dose elagolix group, as compared with 19.6% in the placebo group; in Elaris EM-II, the corresponding percentages were 43.4% and 72.4%, as compared with 22.7% (P
- Published
- 2017
33. Research Priorities for Endometriosis
- Author
-
Rogers, Peter AW, Adamson, G David, Al-Jefout, Moamar, Becker, Christian M, D’Hooghe, Thomas M, Dunselman, Gerard AJ, Fazleabas, Asgerally, Giudice, Linda C, Horne, Andrew W, Hull, M Louise, Hummelshoj, Lone, Missmer, Stacey A, Montgomery, Grant W, Stratton, Pamela, Taylor, Robert N, Rombauts, Luk, Saunders, Philippa T, Vincent, Katy, and Zondervan, Krina T
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Endometriosis ,Contraception/Reproduction ,Consensus ,Education ,Female ,Humans ,Research ,endometriosis ,research priorities ,international workshop ,consensus report ,WES/WERF Consortium for Research Priorities in Endometriosis ,biological marker ,Conference Paper ,disease classification ,embryo development ,epigenetics ,female infertility ,genetics ,genomics ,health care policy ,human ,infertility therapy ,lowest income group ,oocyte ,ovary follicle development ,pain ,pathogenesis ,pathophysiology ,pregnancy outcome ,priority journal ,prognosis ,research priority ,symptom assessment ,workshop ,consensus ,education ,female ,research ,Paediatrics and Reproductive Medicine ,Obstetrics & Reproductive Medicine ,Reproductive medicine ,Midwifery - Abstract
The 3rd International Consensus Workshop on Research Priorities in Endometriosis was held in São Paulo on May 4, 2014, following the 12th World Congress on Endometriosis. The workshop was attended by 60 participants from 19 countries and was divided into 5 main sessions covering pathogenesis/pathophysiology, symptoms, diagnosis/classification/prognosis, disease/symptom management, and research policy. This research priorities consensus statement builds on earlier efforts to develop research directions for endometriosis. Of the 56 research recommendations from the 2011 meeting in Montpellier, a total of 41 remained unchanged, 13 were updated, and 2 were deemed to be completed. Fifty-three new research recommendations were made at the 2014 meeting in Sao Paulo, which in addition to the 13 updated recommendations resulted in a total of 66 new recommendations for research. The research recommendations published herein, as well as those from the 2 previous papers from international consensus workshops, are an attempt to promote high-quality research in endometriosis by identifying and agreeing on key issues that require investigation. New areas included in the 2014 recommendations include infertility, patient stratification, and research in emerging nations, in addition to an increased focus on translational research. A revised and updated set of research priorities that builds on this document will be developed at the 13th World Congress on Endometriosis to be held on May 17-20, 2017, in Vancouver, British Columbia, Canada.
- Published
- 2017
34. World Endometriosis Society consensus on the classification of endometriosis.
- Author
-
Johnson, Neil P, Hummelshoj, Lone, Adamson, G David, Keckstein, Jörg, Taylor, Hugh S, Abrao, Mauricio S, Bush, Deborah, Kiesel, Ludwig, Tamimi, Rulla, Sharpe-Timms, Kathy L, Rombauts, Luk, and Giudice, Linda C
- Subjects
Contraception/Reproduction ,Pain Research ,Clinical Research ,Reproductive health and childbirth ,Good Health and Well Being ,Adult ,Consensus ,Endometriosis ,Female ,Humans ,Quality of Life ,Reproductive Medicine ,endometriosis ,evidence based ,classification ,staging ,World Endometriosis Society Sao Paulo Consortium ,consensus ,r-ASRM ,EFI ,Enzian ,WERF EPHect ,World Endometriosis Society Sao Paulo Consortium ,Medical and Health Sciences ,Studies in Human Society ,Obstetrics & Reproductive Medicine - Abstract
Study questionWhat is the global consensus on the classification of endometriosis that considers the views of women with endometriosis?Summary answerWe have produced an international consensus statement on the classification of endometriosis through systematic appraisal of evidence and a consensus process that included representatives of national and international, medical and non-medical societies, patient organizations, and companies with an interest in endometriosis.What is known alreadyClassification systems of endometriosis, developed by several professional organizations, traditionally have been based on lesion appearance, pelvic adhesions, and anatomic location of disease. One system predicts fertility outcome and none predicts pelvic pain, response to medications, disease recurrence, risks for associated disorders, quality of life measures, and other endpoints important to women and health care providers for guiding appropriate therapeutic options and prognosis.Study design, size, durationA consensus meeting, in conjunction with pre- and post-meeting processes, was undertaken.Participants/materials, setting, methodsA consensus meeting was held on 30 April 2014 in conjunction with the World Endometriosis Society's 12th World Congress on Endometriosis. Rigorous pre- and post-meeting processes, involving 55 representatives of 29 national and international, medical and non-medical organizations from a range of disciplines, led to this consensus statement.Main results and the role of chanceA total of 28 consensus statements were made. Of all, 10 statements had unanimous consensus, however none of the statements was made without expression of a caveat about the strength of the statement or the statement itself. Two statements did not achieve majority consensus. The statements covered women's priorities, aspects of classification, impact of low resources, as well as all the major classification systems for endometriosis. Until better classification systems are developed, we propose a classification toolbox (that includes the revised American Society for Reproductive Medicine and, where appropriate, the Enzian and Endometriosis Fertility Index staging systems), that may be used by all surgeons in each case of surgery undertaken for women with endometriosis. We also propose wider use of the World Endometriosis Research Foundation Endometriosis Phenome and Biobanking Harmonisation Project surgical and clinical data collection tools for research to improve classification of endometriosis in the future, of particular relevance when surgery is not undertaken.Limitations, reasons for cautionThis consensus process differed from that of formal guideline development, although based on the same available evidence. A different group of international experts from those participating in this process may have yielded subtly different consensus statements.Wider implications of the findingsThis is the first time that a large, global, consortium-representing 29 major stake-holding organizations, from 19 countries - has convened to systematically evaluate the best available evidence on the classification of endometriosis and reach consensus. In addition to 21 international medical organizations and companies, representatives from eight national endometriosis organizations were involved, including lay support groups, thus generating and including input from women who suffer from endometriosis in an endeavour to keep uppermost the goal of optimizing quality of life for women with endometriosis.Study funding/competing interestsThe World Endometriosis Society convened and hosted the consensus meeting. Financial support for participants to attend the meeting was provided by the organizations that they represented. There was no other specific funding for this consensus process. Mauricio Abrao is an advisor to Bayer Pharma, and a consultant to AbbVie and AstraZeneca; G David Adamson is the Owner of Advanced Reproductive Care Inc and Ziva and a consultant to Bayer Pharma, Ferring, and AbbVie; Deborah Bush has received travel grants from Fisher & Paykel Healthcare and Bayer Pharmaceuticals; Linda Giudice is a consultant to AbbVie, Juniper Pharmaceutical, and NextGen Jane, holds research grant from the NIH, is site PI on a clinical trial sponsored by Bayer, and is a shareholder in Merck and Pfizer; Lone Hummelshoj is an unpaid consultant to AbbVie; Neil Johnson has received conference expenses from Bayer Pharma, Merck-Serono, and MSD, research funding from AbbVie, and is a consultant to Vifor Pharma and Guerbet; Jörg Keckstein has received a travel grant from AbbVie; Ludwig Kiesel is a consultant to Bayer Pharma, AbbVie, AstraZeneca, Gedeon Richter, and Shionogi, and holds a research grant from Bayer Pharma; Luk Rombauts is an advisor to MSD, Merck Serono, and Ferring, and a shareholder in Monash IVF. The following have declared that they have nothing to disclose: Kathy Sharpe Timms; Rulla Tamimi; Hugh Taylor.Trial registration numberN/A.
- Published
- 2017
35. Insights from imaging the implanting embryo and the uterine environment in three dimensions
- Author
-
Arora, Ripla, Fries, Adam, Oelerich, Karina, Marchuk, Kyle, Sabeur, Khalida, Giudice, Linda C, and Laird, Diana J
- Subjects
Contraception/Reproduction ,2.1 Biological and endogenous factors ,Aetiology ,1.1 Normal biological development and functioning ,Underpinning research ,Reproductive health and childbirth ,Animals ,Blastocyst ,Embryo Implantation ,Embryo ,Mammalian ,Endometrium ,Female ,Humans ,Imaging ,Three-Dimensional ,Mice ,Mice ,Inbred C57BL ,Uterus ,Wnt-5a Protein ,Implantation ,Receptivity ,Embryo ,Confocal Imaging ,Surface curvature ,Wnt5a ,Biological Sciences ,Medical and Health Sciences - Abstract
Although much is known about the embryo during implantation, the architecture of the uterine environment in which the early embryo develops is not well understood. We employed confocal imaging in combination with 3D analysis to identify and quantify dynamic changes to the luminal structure of murine uterus in preparation for implantation. When applied to mouse mutants with known implantation defects, this method detected striking peri-implantation abnormalities in uterine morphology that cannot be visualized by histology. We revealed 3D organization of uterine glands and found that they undergo a stereotypical reorientation concurrent with implantation. Furthermore, we extended this technique to generate a 3D rendering of the cycling human endometrium. Analyzing the uterine and embryo structure in 3D for different genetic mutants and pathological conditions will help uncover novel molecular pathways and global structural changes that contribute to successful implantation of an embryo.
- Published
- 2016
36. Aberrant Endometrial DNA Methylome and Associated Gene Expression in Women with Endometriosis1
- Author
-
Houshdaran, Sahar, Nezhat, Camran R, Vo, Kim Chi, Zelenko, Zara, Irwin, Juan C, and Giudice, Linda C
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Genetics ,Biological Sciences ,Clinical Research ,Endometriosis ,Pain Research ,Contraception/Reproduction ,Infertility ,Biotechnology ,Aetiology ,2.1 Biological and endogenous factors ,Underpinning research ,1.1 Normal biological development and functioning ,Reproductive health and childbirth ,Adult ,CpG Islands ,DNA Methylation ,Endometrium ,Epigenesis ,Genetic ,Female ,Gene Expression ,Gene Expression Regulation ,Humans ,Menstrual Cycle ,Proteomics ,DNA methylation ,endometriosis ,endometrium ,epigenetics ,Medical and Health Sciences ,Obstetrics & Reproductive Medicine ,Animal production ,Zoology ,Reproductive medicine - Abstract
Endometriosis is an estrogen-dependent, progesterone-resistant disorder largely derived from retrograde transplantation of menstrual tissue/cells into the pelvis, eliciting an inflammatory response, pelvic pain, and infertility. Eutopic endometrium (within the uterus), giving rise to pelvic disease, displays cycle-dependent transcriptomic, proteomic, and signaling abnormalities, and although its DNA methylation profiles dynamically change across the cycle in healthy women, studies in endometriosis are limited. Herein, we investigated the DNA methylome and associated gene expression in three phases of the cycle in eutopic endometrium of women with severe endometriosis versus controls, matched for ethnicity, medications, smoking, and no recent contraceptive steroid use. Genome-wide DNA methylation and gene expression were coassessed in each sample. Cycle phase was determined by histology, serum hormone levels, and unsupervised principal component and hierarchical cluster analyses of microarray data. Altered endometrial DNA methylation in endometriosis was most prominent in the midsecretory phase (peak progesterone), with disruption of the normal pattern of cycle-dependent DNA methylation changes, including a bias toward methylation of CpG islands, suggesting wide-range abnormalities of the chromatin remodeling machinery in endometriosis. DNA methylation changes were associated with altered gene expression relevant to endometrial function/dysfunction, including cell proliferation, inflammation/immune response, angiogenesis, and steroid hormone response. The data provide insight into epigenetic reprogramming and steroid hormone actions in endometrium contributing to the pathogenesis and pathophysiology of endometriosis.
- Published
- 2016
37. Global Transcriptome Abnormalities of the Eutopic Endometrium From Women With Adenomyosis
- Author
-
Herndon, Christopher N, Aghajanova, Lusine, Balayan, Shaina, Erikson, David, Barragan, Fatima, Goldfien, Gabriel, Vo, Kim Chi, Hawkins, Shannon, and Giudice, Linda C
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Clinical Research ,Genetics ,Contraception/Reproduction ,Endometriosis ,Aetiology ,2.1 Biological and endogenous factors ,Adenomyosis ,Adult ,Cell Proliferation ,Endometrium ,Female ,Gene Expression Profiling ,Humans ,Myometrium ,Transcriptome ,adenomyosis ,eutopic endometrium ,microarray ,apoptosis ,signaling pathways ,Paediatrics and Reproductive Medicine ,Obstetrics & Reproductive Medicine ,Reproductive medicine ,Midwifery - Abstract
ObjectiveAdenomyosis is a clinical disorder defined by the presence of endometrial glands and stroma within the myometrium, the pathogenesis of which is poorly understood. We postulate that dysregulation of genes and pathways in eutopic endometrium may predispose to ectopic implantation. No study, to our knowledge, has examined the global transcriptome of isolated eutopic endometrium from women with clinically significant adenomyosis.DesignLaboratory-based study with full institutional review board approval and consents.Material and methodsEndometrial sampling was performed on hysterectomy specimens (proliferative phase) from symptomatic women with pathologically confirmed diffuse adenomyosis (n = 3). Controls (n = 5) were normo-ovulatory patients without adenomyosis. All patients were free from leiomyoma, endometriosis, and hormonal exposures. Isolated purified total RNA was subjected to microarray analysis using the Gene 1.0 ST Affymetrix platform. Data were analyzed with GeneSpring and Ingenuity Pathway analysis. Validation of several genes was undertaken by quantitative real-time reverse transcriptase polymerase chain reaction.ResultsComparison of transcriptomes of proliferative endometrium from women with and without adenomyosis revealed 140 upregulated and 884 downregulated genes in samples from women with adenomyosis compared to controls. Highly differentially expressed genes include those involved in regulation of apoptosis, steroid hormone responsiveness, and proteins involved in extracellular matrix remodeling as well as microRNAs of unknown significance. Affected canonical pathways included eukaryotic initiation factor 2 signaling, oxidative phosphorylation, mitochondrial dysfunction, estrogen receptor signaling, and mammalian target of rapamycin signaling.ConclusionThe eutopic endometrium in patients with adenomyosis has fundamental abnormalities that may predispose to invasion and survival beyond the myometrial interface.
- Published
- 2016
38. Effects of the levonorgestrel‐releasing intrauterine device on the immune microenvironment of the human cervix and endometrium
- Author
-
Shanmugasundaram, Uma, Hilton, Joan F, Critchfield, J William, Greenblatt, Ruth M, Giudice, Linda C, Averbach, Sarah, Seidman, Dominika, Shacklett, Barbara L, and Smith‐McCune, Karen
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Immunology ,HIV/AIDS ,Prevention ,Women's Health ,Contraception/Reproduction ,Sexually Transmitted Infections ,Infectious Diseases ,Clinical Research ,Adult ,Cervix Uteri ,Cross-Sectional Studies ,Drug Delivery Systems ,Endometrium ,Female ,HIV Infections ,HLA-DR Antigens ,Humans ,Interleukin-10 ,Levonorgestrel ,Middle Aged ,Receptors ,CCR4 ,Receptors ,CCR5 ,Risk Factors ,T-Lymphocytes ,chemokine ,cytokine ,HIV ,IUD ,progestin ,T-cell - Abstract
ProblemThere is little information regarding the impact of the intrauterine device on immune parameters of the upper female reproductive tract related to risk of HIV acquisition.Method of studyWe collected cervical and endometrial samples from women using the hormonal intrauterine device to study its effects on endocervical cytokines/chemokine concentrations, phenotypic markers of T cells, responses of endometrial T cells to activation, and alterations of endometrial cellular infiltrates.ResultsHormonal intrauterine device use was associated with: increased concentrations of inflammatory cytokines/chemokines (endocervix); increased coexpression of CXCR4 and CCR5 (endocervix and endometrium); increased coexpression of CD38 and HLADR (endocervix and endometrium); increased intracellular IL-10 production after T-cell stimulation (endometrium); and increased density of T cells, most notably regulatory T cells (endometrium).ConclusionHormonal intrauterine device use resulted in both inflammatory and immunosuppressive alterations. Further research is needed to determine the significance of these changes for HIV risk.
- Published
- 2016
39. Moving from awareness to action on preventing patient exposure to toxic environmental chemicals
- Author
-
Sutton, Patrice M, Giudice, Linda C, and Woodruff, Tracey J
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Environment ,Environmental Exposure ,Environmental Pollutants ,Humans ,Paediatrics and Reproductive Medicine ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Published
- 2016
40. Human Endometrial Fibroblasts Derived from Mesenchymal Progenitors Inherit Progesterone Resistance and Acquire an Inflammatory Phenotype in the Endometrial Niche in Endometriosis1
- Author
-
Barragan, Fatima, Irwin, Juan C, Balayan, Shaina, Erikson, David W, Chen, Joseph C, Houshdaran, Sahar, Piltonen, Terhi T, Spitzer, Trimble LB, George, Ashley, Rabban, Joseph T, Nezhat, Camran, and Giudice, Linda C
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Stem Cell Research - Nonembryonic - Human ,Regenerative Medicine ,Stem Cell Research - Nonembryonic - Non-Human ,Genetics ,Clinical Research ,Endometriosis ,Stem Cell Research ,1.1 Normal biological development and functioning ,Underpinning research ,Aetiology ,2.1 Biological and endogenous factors ,Case-Control Studies ,Cell Differentiation ,Cell Proliferation ,Cells ,Cultured ,Endometrium ,Female ,Fibroblasts ,Humans ,Inflammation ,Inflammation Mediators ,Mesenchymal Stem Cells ,Phenotype ,Stem Cell Niche ,Transcriptome ,Uterine Diseases ,differentiation ,endometriosis ,endometrium ,fibroblasts ,mesenchymal stem cells ,Biological Sciences ,Medical and Health Sciences ,Obstetrics & Reproductive Medicine ,Animal production ,Zoology ,Reproductive medicine - Abstract
Human endometrium undergoes cyclic regeneration involving stem/progenitor cells, but the role of resident endometrial mesenchymal stem cells (eMSC) as progenitors of endometrial stromal fibroblasts (eSF) has not been definitively demonstrated. In endometriosis, eSF display progesterone (P4) resistance with impaired decidualization in vivo and in vitro. To investigate eMSC as precursors of eSF and whether endometriosis P4 resistance is inherited from eMSC, we analyzed transcriptomes of eutopic endometrium eMSC and eSF isolated by fluorescence-activated cell sorting (FACS) from endometriosis (eMSCendo, eSFendo) and controls (eMSCcontrol, eSFcontrol) and their derived primary cultures. Differentially expressed lineage-associated genes (LG) of FACS-isolated eMSC and eSF were largely conserved in endometriosis. In culture, eSFcontrol maintained in vitro expression of a subset of eSF LG and decidualized in vitro with P4 The eMSCcontrol cultures differentiated in vitro to eSF lineage, down-regulating eMSC LG and up-regulating eSF LG, showing minimal transcriptome differences versus eSFcontrol cultures and decidualizing in vitro. Cultured eSFendo displayed less in vitro LG stability and did not decidualize in vitro. In vitro, eMSCendo differentiated to eSF lineage but showed more differentially expressed genes versus eSFendo cultures, and did not decidualize in vitro, demonstrating P4 resistance inherited from eMSCendo Compared to controls, cultures from tissue-derived eSFendo uniquely had a pro-inflammatory phenotype not present in eMSCendo differentiated to eSF in vitro, suggesting divergent niche effects for in vivo versus in vitro lineage differentiation. These findings substantiate eMSC as progenitors of eSF and reveal eSF in endometriosis as having P4 resistance inherited from eMSC and a pro-inflammatory phenotype acquired within the endometrial niche.
- Published
- 2016
41. Cryopreservation and recovery of human endometrial epithelial cells with high viability, purity, and functional fidelity
- Author
-
Chen, Joseph C, Hoffman, Jacquelyn R, Arora, Ripla, Perrone, Lila A, Gonzalez-Gomez, Christian J, Vo, Kim Chi, Laird, Diana J, Irwin, Juan C, and Giudice, Linda C
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Uterine Cancer ,Clinical Research ,Cancer ,Biomarkers ,Cell Polarity ,Cell Separation ,Cell Shape ,Cell Survival ,Cells ,Cultured ,Cryopreservation ,Endometrium ,Epithelial Cells ,Female ,Gene Expression Regulation ,Humans ,Permeability ,Phenotype ,Tight Junctions ,freezing medium ,endometrium ,endometrial epithelium ,endometrial epithelial cells ,Clinical Sciences ,Paediatrics and Reproductive Medicine ,Public Health and Health Services ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Abstract
ObjectiveTo develop a protocol for cryopreservation and recovery of human endometrial epithelial cells (eECs) retaining molecular and functional characteristics of endometrial epithelium in vivo.DesignIn vitro study using human endometrial cells.SettingUniversity research laboratory.Patient(s)Endometrial biopsies were obtained from premenopausal women undergoing benign gynecologic procedures.Intervention(s)Primary eECs were cryopreserved in 1% fetal bovine serum/10% dimethylsulfoxide in Defined Keratinocyte Serum-Free Medium (KSFM). Recovered cells were observed for endometrial stromal fibroblast (eSF) contamination and subsequently evaluated for morphology, gene expression, and functional characteristics of freshly cultured eECs and in vivo endometrial epithelium.Main outcome measure(s)Analysis of eEC morphology and the absence of eSF contamination; evaluation of epithelial-specific gene and protein expression; assessment of epithelial polarity.Result(s)Endometrial epithelial cells recovered after cryopreservation (n = 5) displayed epithelial morphology and expressed E-cadherin (CDH1), occludin (OCLN), claudin1 (CLDN1), and keratin18 (KRT18). Compared with eSF, recovered eECs displayed increased (P
- Published
- 2016
42. Infertility and reproductive disorders: impact of hormonal and inflammatory mechanisms on pregnancy outcome
- Author
-
Vannuccini, Silvia, Clifton, Vicki L, Fraser, Ian S, Taylor, Hugh S, Critchley, Hilary, Giudice, Linda C, and Petraglia, Felice
- Subjects
Prevention ,Infertility ,Infant Mortality ,Perinatal Period - Conditions Originating in Perinatal Period ,Pediatric ,Preterm ,Low Birth Weight and Health of the Newborn ,Contraception/Reproduction ,Aetiology ,2.1 Biological and endogenous factors ,Reproductive health and childbirth ,Good Health and Well Being ,Cervix Uteri ,Female ,Female Urogenital Diseases ,Gonadal Steroid Hormones ,Humans ,Infertility ,Female ,Inflammation ,Leiomyoma ,Maternal Age ,Placenta ,Polycystic Ovary Syndrome ,Pregnancy ,Pregnancy Complications ,Pregnancy Outcome ,Premature Birth ,Reproductive Techniques ,Assisted ,Signal Transduction ,polycystic ovary syndrome ,endometriosis ,uterine fibroids ,unexplained infertility ,assisted reproductive technologies ,preterm birth ,pre-eclampsia ,placenta ,inflammation ,sex steroids ,Clinical Sciences ,Paediatrics and Reproductive Medicine ,Public Health and Health Services ,Obstetrics & Reproductive Medicine - Abstract
BackgroundReproductive disorders and infertility are associated with the risk of obstetric complications and have a negative impact on pregnancy outcome. Affected patients often require assisted reproductive technologies (ART) to conceive, and advanced maternal age is a further confounding factor. The challenge is to dissect causation, correlation and confounders in determining how infertility and reproductive disorders individually or together predispose women to poor pregnancy outcomes.MethodsThe published literature, to June 2015, was searched using PubMed, summarizing all evidences concerning the perinatal outcome of women with infertility and reproductive disorders and the potential mechanisms that may influence poor pregnancy outcome.ResultsReproductive disorders (endometriosis, adenomyosis, polycystic ovary syndrome and uterine fibroids) and unexplained infertility share inflammatory pathways, hormonal aberrations, decidual senescence and vascular abnormalities that may impair pregnancy success through common mechanisms. Either in combination or alone, these disorders results in an increased risk of preterm birth, fetal growth restriction, placental pathologies and hypertensive disorders. Systemic hormonal aberrations, and inflammatory and metabolic factors acting on endometrium, myometrium, cervix and placenta are all associated with an aberrant milieu during implantation and pregnancy, thus contributing to the genesis of obstetric complications. Some of these features have been also described in placentas from ART.ConclusionsReproductive disorders are common in women of childbearing age and rarely occur in isolation. Inflammatory, endocrine and metabolic mechanisms associated with these disorders are responsible for an increased incidence of obstetric complications. These patients should be recognized as 'high risk' for poor pregnancy outcomes and monitored with specialized follow-up. There is a real need for development of evidence-based recommendations about clinical management and specific obstetric care pathways for the introduction of prompt preventative care measures.
- Published
- 2016
43. Manufacturing doubt about endocrine disrupter science--A rebuttal of industry-sponsored critical comments on the UNEP/WHO report "State of the Science of Endocrine Disrupting Chemicals 2012".
- Author
-
Bergman, Åke, Becher, Georg, Blumberg, Bruce, Bjerregaard, Poul, Bornman, Riana, Brandt, Ingvar, Casey, Stephanie C, Frouin, Heloise, Giudice, Linda C, Heindel, Jerrold J, Iguchi, Taisen, Jobling, Susan, Kidd, Karen A, Kortenkamp, Andreas, Lind, P Monica, Muir, Derek, Ochieng, Roseline, Ropstad, Erik, Ross, Peter S, Skakkebaek, Niels Erik, Toppari, Jorma, Vandenberg, Laura N, Woodruff, Tracey J, and Zoeller, R Thomas
- Subjects
Animals ,Humans ,Endocrine Disruptors ,EDCs ,Endocrine disruption ,Endocrine disruptors ,Pharmacology and Pharmaceutical Sciences ,Toxicology - Abstract
We present a detailed response to the critique of "State of the Science of Endocrine Disrupting Chemicals 2012" (UNEP/WHO, 2013) by financial stakeholders, authored by Lamb et al. (2014). Lamb et al.'s claim that UNEP/WHO (2013) does not provide a balanced perspective on endocrine disruption is based on incomplete and misleading quoting of the report through omission of qualifying statements and inaccurate description of study objectives, results and conclusions. Lamb et al. define extremely narrow standards for synthesizing evidence which are then used to dismiss the UNEP/WHO 2013 report as flawed. We show that Lamb et al. misuse conceptual frameworks for assessing causality, especially the Bradford-Hill criteria, by ignoring the fundamental problems that exist with inferring causality from empirical observations. We conclude that Lamb et al.'s attempt of deconstructing the UNEP/WHO (2013) report is not particularly erudite and that their critique is not intended to be convincing to the scientific community, but to confuse the scientific data. Consequently, it promotes misinterpretation of the UNEP/WHO (2013) report by non-specialists, bureaucrats, politicians and other decision makers not intimately familiar with the topic of endocrine disruption and therefore susceptible to false generalizations of bias and subjectivity.
- Published
- 2015
44. International Federation of Gynecology and Obstetrics opinion on reproductive health impacts of exposure to toxic environmental chemicals
- Author
-
Di Renzo, Gian Carlo, Conry, Jeanne A, Blake, Jennifer, DeFrancesco, Mark S, DeNicola, Nathaniel, Martin, James N, McCue, Kelly A, Richmond, David, Shah, Abid, Sutton, Patrice, Woodruff, Tracey J, van der Poel, Sheryl Ziemin, and Giudice, Linda C
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Contraception/Reproduction ,Nutrition ,Patient Safety ,Prevention ,2.1 Biological and endogenous factors ,Aetiology ,2.2 Factors relating to the physical environment ,Reproductive health and childbirth ,Good Health and Well Being ,Breast Feeding ,Cost of Illness ,Environmental Exposure ,Environmental Monitoring ,Environmental Pollutants ,Female ,Global Health ,Humans ,International Agencies ,Pregnancy ,Prenatal Exposure Delayed Effects ,Reproduction ,Risk Factors ,Socioeconomic Factors ,Developmental health ,Environmental chemicals ,Reproductive environmental health ,Toxic chemicals ,Women's health ,Paediatrics and Reproductive Medicine ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Abstract
Exposure to toxic environmental chemicals during pregnancy and breastfeeding is ubiquitous and is a threat to healthy human reproduction. There are tens of thousands of chemicals in global commerce, and even small exposures to toxic chemicals during pregnancy can trigger adverse health consequences. Exposure to toxic environmental chemicals and related health outcomes are inequitably distributed within and between countries; universally, the consequences of exposure are disproportionately borne by people with low incomes. Discrimination, other social factors, economic factors, and occupation impact risk of exposure and harm. Documented links between prenatal exposure to environmental chemicals and adverse health outcomes span the life course and include impacts on fertility and pregnancy, neurodevelopment, and cancer. The global health and economic burden related to toxic environmental chemicals is in excess of millions of deaths and billions of dollars every year. On the basis of accumulating robust evidence of exposures and adverse health impacts related to toxic environmental chemicals, the International Federation of Gynecology and Obstetrics (FIGO) joins other leading reproductive health professional societies in calling for timely action to prevent harm. FIGO recommends that reproductive and other health professionals advocate for policies to prevent exposure to toxic environmental chemicals, work to ensure a healthy food system for all, make environmental health part of health care, and champion environmental justice.
- Published
- 2015
45. Reducing low-value care in endometriosis between limited evidence and unresolved issues: a proposal
- Author
-
Vercellini, Paolo, Giudice, Linda C, Evers, Johannes LH, and Abrao, Mauricio S
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Contraception/Reproduction ,Endometriosis ,Prevention ,Patient Safety ,Pain Research ,Health Services ,Comparative Effectiveness Research ,Clinical Research ,Reproductive health and childbirth ,Female ,Humans ,Practice Guidelines as Topic ,Quality of Health Care ,endometriosis ,infertility ,pelvic pain ,patient-centred medicine ,low-value care ,Medical and Health Sciences ,Studies in Human Society ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Abstract
Quantification of benefits and harms of medical interventions should be based on high-quality evidence, which is not always the case in the endometriosis field. In many clinical circumstances, healthcare decisions in women with endometriosis are taken based on suboptimal evidence or on evidence of coexistence of benefits and harms that must be balanced. In these conditions, it is important to avoid or reduce the use of low-value care, i.e. interventions with defined harms and uncertain benefits, or whose effectiveness is comparable with less expensive alternatives. In particular, we suggest that: (i) non-surgical diagnosis based on symptoms, physical findings and transvaginal ultrasonography is possible in most women with symptomatic endometriosis. Thus, except in doubtful cases, laparoscopy should be intended for surgical treatment, not for diagnostic purposes: early diagnosis and diagnostic laparoscopy are not synonymous; (ii) future trials on new drugs for endometriosis should address those outcomes that are most important to patients, should be designed as superiority trials and should include a progestin or an estrogen-progestin as a comparator. Moreover, limitation of repetitive surgery for recurrent endometriosis is among the objectives of long-term medical treatment; (iii) indications for surgery should be the result of a balance between demonstrated benefits in terms of fertility enhancement and pain relief, specific risks associated with excision of different types of endometriotic lesions, cost-effectiveness and patient preference after detailed information; (iv) physicians, health professionals and policy makers should discriminate between screening for and diagnosis of endometriosis. Limited peritoneal foci, which are frequently observed also in asymptomatic women, regress or remain stable in about two thirds of cases. Therefore, the theoretical premises for a screening campaign are currently unclear; (v) physicians should develop the ability to effectively communicate quantitative information based on international guidelines and systematic literature reviews. This will assist a woman's understanding of the interaction between the evidence and her priorities, facilitating the transition towards value-based medicine.
- Published
- 2015
46. Progestin-Containing Contraceptives Alter Expression of Host Defense-Related Genes of the Endometrium and Cervix.
- Author
-
Goldfien, Gabriel A, Barragan, Fatima, Chen, Joseph, Takeda, Margaret, Irwin, Juan C, Perry, Jean, Greenblatt, Ruth M, Smith-McCune, Karen K, and Giudice, Linda C
- Subjects
Cervix Uteri ,Endometrium ,Humans ,Levonorgestrel ,Contraceptive Agents ,Female ,Progestins ,Cross-Sectional Studies ,Gene Expression Regulation ,Adolescent ,Adult ,Female ,Young Adult ,Medroxyprogesterone Acetate ,cervix ,endometrium ,gene expression ,host defense ,progestin-based contraceptives ,HIV/AIDS ,Genetics ,Infectious Diseases ,Contraception/Reproduction ,Clinical Research ,2.1 Biological and endogenous factors ,Obstetrics & Reproductive Medicine ,Paediatrics and Reproductive Medicine - Abstract
Epidemiological studies indicate that progestin-containing contraceptives increase susceptibility to HIV, although the underlying mechanisms involving the upper female reproductive tract are undefined. To determine the effects of depot medroxyprogesterone acetate (DMPA) and the levonorgestrel intrauterine system (LNG-IUS) on gene expression and physiology of human endometrial and cervical transformation zone (TZ), microarray analyses were performed on whole tissue biopsies. In endometrium, activated pathways included leukocyte chemotaxis, attachment, and inflammation in DMPA and LNG-IUS users, and individual genes included pattern recognition receptors, complement components, and other immune mediators. In cervical TZ, progestin treatment altered expression of tissue remodeling and viability but not immune function genes. Together, these results indicate that progestins influence expression of immune-related genes in endometrium relevant to local recruitment of HIV target cells with potential to increase susceptibility and underscore the importance of the upper reproductive tract when assessing the safety of contraceptive products.
- Published
- 2015
47. Krüppel-Like Factor 13 Deficiency in Uterine Endometrial Cells Contributes to Defective Steroid Hormone Receptor Signaling but Not Lesion Establishment in a Mouse Model of Endometriosis.
- Author
-
Heard, Melissa E, Velarde, Michael C, Giudice, Linda C, Simmen, Frank A, and Simmen, Rosalia CM
- Subjects
Endometrium ,Animals ,Mice ,Knockout ,Humans ,Mice ,Endometriosis ,Disease Models ,Animal ,Estradiol ,Progesterone ,Cell Cycle Proteins ,Receptors ,Estrogen ,Receptors ,Progesterone ,Repressor Proteins ,Signal Transduction ,Apoptosis ,Cell Differentiation ,Female ,Kruppel-Like Transcription Factors ,Krüppel-like factor 13 ,Krüppel-like factor 9 ,endometriosis ,estrogen receptor ,mouse model ,progesterone receptor ,Obstetrics & Reproductive Medicine ,Medical and Health Sciences ,Biological Sciences - Abstract
Krüppel-like Factor (KLF) 13 and the closely related KLF9 are members of the Sp/KLF family of transcription factors that have collectively emerged as essential regulators of tissue development, differentiation, proliferation, and programmed cell death. Steroid hormone-responsive tissues express multiple KLFs that are linked to progesterone receptor (PGR) and estrogen receptor (ESR) actions either as integrators or as coregulators. Endometriosis is a chronic disease characterized by progesterone resistance and dysregulated estradiol signaling; nevertheless, distinct KLF members' contributions to endometriosis remain largely undefined. We previously demonstrated promotion of ectopic lesion establishment by Klf9 null endometrium in a mouse model of endometriosis. Here we evaluated whether KLF13 loss of expression in endometrial cells may equally contribute to lesion formation. KLF13 transcript levels were lower in the eutopic endometria of women with versus women without endometriosis at menstrual midsecretory phase. In wild-type (WT) mouse recipients intraperitoneally administered WT or Klf13 null endometrial fragments, lesion incidence did not differ with donor genotype. No differences were noted for lesion volume, number, proliferation status, and apoptotic index as well. Klf13 null lesions displayed reduced total PGR and ESR1 (RNA and immunoreactive protein) and altered expression of several PGR and ESR1 target genes, relative to WT lesions. Unlike for Klf9 null lesions, changes in transcript levels for PGR-A, ESR1, and Notch/Hedgehog-associated pathway components were not observed for Klf13 null lesions. Results demonstrate lack of a causative relationship between endometrial KLF13 deficiency and lesion establishment in mice, and they support the broader participation of multiple signaling pathways, besides those mediated by steroid receptors, in the pathology of endometriosis.
- Published
- 2015
48. Endometrial stromal fibroblasts from women with polycystic ovary syndrome have impaired progesterone-mediated decidualization, aberrant cytokine profiles and promote enhanced immune cell migration in vitro
- Author
-
Piltonen, TT, Chen, JC, Khatun, M, Kangasniemi, M, Liakka, A, Spitzer, T, Tran, N, Huddleston, H, Irwin, JC, and Giudice, LC
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Contraception/Reproduction ,Clinical Research ,Infertility ,2.1 Biological and endogenous factors ,Aetiology ,Reproductive health and childbirth ,Adult ,Biopsy ,Case-Control Studies ,Cell Movement ,Cell Proliferation ,Cytokines ,Decidua ,Embryo Implantation ,Endometrial Neoplasms ,Endometrium ,Estrogens ,Female ,Fibroblasts ,Gene Expression Regulation ,Genetic Predisposition to Disease ,Glucose Tolerance Test ,Humans ,Middle Aged ,Polycystic Ovary Syndrome ,Pregnancy ,Progesterone ,Prospective Studies ,PCOS ,endometrium ,decidualization ,cytokines ,MMP ,Medical and Health Sciences ,Studies in Human Society ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Abstract
Study questionDo endometrial stromal fibroblasts (eSF) in women with polycystic ovary syndrome (PCOS) (eSFpcos) exhibit altered estrogen and/or progesterone (P4) responses, which may explain some of the adverse reproductive outcomes and endometrial pathologies in these women?Summary answerIn vitro, eSF from women with PCOS exhibit an aberrant decidualization response and concomitant changes in pro-inflammatory cytokine, chemokine and matrix metalloproteinase (MMP) release and immune cell chemoattraction. In vivo these aberrations may result in suboptimal implantation and predisposition to endometrial cancer.What is known alreadyThe endometrium in women with PCOS has several abnormalities including progesterone (P4) resistance at the gene expression level, likely contributing to subfertility, pregnancy complications and increased endometrial cancer risk in PCOS women.Study design, size, durationProspective, university-based, case-control, in vitro study.Participants/materials, setting, methodsCultures of eSFPCOS (n = 12, Rotterdam and NIH criteria) and eSFControl (Ctrl) (n = 6, regular cycle length, no signs of hyperandrogenism) were treated with vehicle, estradiol (E2, 10 nM) or E2P4 (10 nM/1 μM) for 14 days. Progesterone receptor (PGR) mRNA was assessed with quantitative real-time PCR (qRT-PCR) and eSF decidualization was confirmed by insulin-like growth factor-binding protein-1 (IGFBP-1) transcript and protein expression. Fractalkine (CX3CL1), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin (IL) 6, 8 and 11, macrophage chemoattractant protein (MCP) 1 and 3, CCL5 (RANTES) and MMPs (MMP1, 2, 3, 7, 9, 10 and 12) were measured in conditioned media by Luminex multiplex assays, and chemotactic activity of the conditioned media was tested in a migration assay using CD14+ monocyte and CD4+ T-cell migration assay. Effects of IL-6 (0.02, 0.2, 2 or 20 ng/ml) or IL-8 (0.04, 0.4, 4, or 40 ng/ml) or combination (0.2 ng/ml IL-6 and 4.0 ng/ml IL-8) on 14-d decidualization were also tested. ANOVA with pre-planned contrasts was used for statistical analysis.Main results and the role of chanceHormonal challenge with E2P4 to induce decidualization revealed two distinct subsets of eSFPCOS. Eight eSFPCOS (dPCOS) and all eSFCtrl (dCtrl) cultures showed a normal decidualization response to E2P4 as determined by morphology and IGFBP-1 secretion. However, 4 eSFPCOS cultures showed blunted decidualization (ndPCOS) in morphological assessment and low IGFBP-1 levels even though all three groups exhibited normal estrogen-mediated increase in PGR expression. Interestingly dPCOS had decreased IL-6 and GM-SCF secretion compared with dCtrl, whereas the ndPCOS cultures showed increased IL-6 and 8, MCP1, RANTES and GM-CSF secretion at base-line and/or in response to E2 or E2P4 compared with dCtrl and/or dPCOS. Furthermore, even though PGR expression was similar in all three groups, P4 inhibition of MMP secretion was attenuated in ndPCOS resulting in higher MMP2 and 3 levels. The conditioned media from ndPCOS had increased chemoattractic activity compared with dCtrl and dPCOS media. Exogenously added IL-6 and/or 8 did not inhibit decidualization in eSFCtrl indicating that high levels of these cytokines in ndPCOS samples were not likely a cause for the aberrant decidualization.Limitations, reasons for cautionThis is an in vitro study with a small sample size, utilizing stromal cell cultures from proliferative and secretory phase endometrium. The effect of PCOS on endometrial epithelium, another major histoarchitectural cell compartment of the endometrium, was not evaluated and should be considered in future studies. Furthermore, results obtained should also be confirmed in a larger data set and with mid/late secretory phase in vivo samples and models.Wider implications of the findingsThe alterations seen in ndPCOS may contribute to endometrial dysfunction, subfertility and pregnancy complications in PCOS women. The results emphasize the importance of understanding immune responses related to the implantation process and normal endometrial homeostasis in women with PCOS.Study funding/competing interestsSigrid Juselius Foundation, Academy of Finland, Finnish Medical Foundation, Orion-Farmos Research Foundation (to T.T.P.), the NIH Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) U54HD 055764-07 Specialized Cooperative Centers Program in Reproduction and Infertility Research (to L.C.G.), the NICHD the Ruth L. Kirschstein National Research Service Awards grant 1F32HD074423-03 (to J.C.C.). The authors have no competing interests.
- Published
- 2015
49. Urine, peritoneal fluid and omental fat proteomes of reproductive age women: Endometriosis-related changes and associations with endocrine disrupting chemicals
- Author
-
Williams, Katherine E, Miroshnychenko, Olga, Johansen, Eric B, Niles, Richard K, Sundaram, Rajeshwari, Kannan, Kurunthachalam, Albertolle, Matthew, Zhou, Yan, Prasad, Namrata, Drake, Penelope M, Giudice, Linda C, Hall, Steven C, Witkowska, H Ewa, Louis, Germaine M Buck, and Fisher, Susan J
- Subjects
Reproductive Medicine ,Biomedical and Clinical Sciences ,Endometriosis ,Estrogen ,Infertility ,Contraception/Reproduction ,Clinical Research ,Women's Health ,Pain Research ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Reproductive health and childbirth ,Good Health and Well Being ,Adipose Tissue ,Adult ,Ascitic Fluid ,Contraceptives ,Oral ,Hormonal ,Female ,Humans ,Leukocyte Elastase ,Matrix Metalloproteinase 9 ,Omentum ,Proteome ,Peritoneal fluid ,Urine ,Omental fat ,Mass spectrometry ,Environmental chemicals ,Biochemistry and Cell Biology ,Plant Biology ,Analytical Chemistry ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Plant biology ,Medical biochemistry and metabolomics - Abstract
Endometriosis, ectopic growth of the uterine lining (endometrium), which affects 6-11% of reproductive age women, is associated with pelvic pain and infertility. We investigated the peritoneal fluid (PF), urine and omental fat (OF) proteomes of women with endometriosis vs. individuals with no surgically visualized endometriosis. All participants were enrolled in the NICHD-funded ENDO Study. A two-step proteomic study was performed. The first, a broad survey, employed a semi-quantitative gel LC-mass spectrometry (MS) workflow: SDS PAGE fractionation, trypsin digestion and LC-MS/MS. The results showed sample integrity but failed to detect any differences between women with and without endometriosis. The second step was a quantitative analysis of OF samples. We employed another sample set (n=30) from women ± disease and isobaric mass-tag (iTRAQ) chemistry to label peptides and 2D LC-MS/MS for protein identification and quantification. Three proteins-matrix metalloproteinase-9, neutrophil elastase, and FAM49B-were significantly lower in abundance in samples from women with endometriosis. Interestingly, neutrophil elastase and FAM49B levels were associated with higher levels of a subset of endocrine disrupting chemicals (EDCs) that were previously measured in the same samples. The results of these experiments showed the feasibility of associating endometriosis with changes in the OF protein repertoire and EDC levels.Biological significanceEndometriosis, pathological growth of the uterine lining, is associated with significant morbidities, including pain and infertility. However, the causes of this common condition are poorly understood. This study determined whether endometriosis was associated with changes in the protein composition of peritoneal fluid, urine and/or omental fat. A protein of unknown function (FAM49B) and two proteinases (metalloproteinase-9, neutrophil elastase) were down regulated in OF samples from women with versus without endometriosis. These findings suggested proteinase imbalances at sites that were distant from the endometriotic lesions. Additionally, FAM49B and neutrophil elastase levels were associated with higher levels of a subset of environmental chemicals that were quantified in the same samples, suggesting other possible associations. Thus, this work generated hypotheses that will be tested in further studies.
- Published
- 2015
50. Unexpected Inflammatory Effects of Intravaginal Gels (Universal Placebo Gel and Nonoxynol-9) on the Upper Female Reproductive Tract: A Randomized Crossover Study.
- Author
-
Smith-McCune, Karen, Chen, Joseph C, Greenblatt, Ruth M, Shanmugasundaram, Uma, Shacklett, Barbara L, Hilton, Joan F, Johnson, Brittni, Irwin, Juan C, and Giudice, Linda C
- Subjects
Uterus ,Cervix Uteri ,T-Lymphocytes ,Humans ,Inflammation ,Nonoxynol ,Gels ,Placebos ,Anti-HIV Agents ,Administration ,Intravaginal ,Cross-Over Studies ,Phenotype ,Adult ,Female ,Young Adult ,Transcriptome ,Cellular Microenvironment ,General Science & Technology - Abstract
Intravaginal anti-HIV microbicides could provide women with a self-controlled means for HIV prevention, but results from clinical trials have been largely disappointing. We postulated that unrecognized effects of intravaginal gels on the upper female reproductive tract might contribute to the lower-than-expected efficacy of HIV microbicides. Our objective was to study the effects of intravaginal gels on the immune microenvironment of the cervix and uterus. In this randomized crossover study, 27 healthy female volunteers used a nightly application of intravaginal nonoxynol-9 (N9) gel as a "failed" microbicide or the universal placebo gel (UPG) as a "safe" gel (intervention cycles), or nothing (control cycle) from the end of menses to the mid-luteal phase. At a specific time-point following ovulation, all participants underwent sample collection for measurements of T-cell phenotypes, gene expression, and cytokine/chemokine protein concentrations from 3 anatomic sites above the vagina: the cervical transformation zone, the endocervix and the endometrium. We used hierarchical statistical models to estimate mean (95% CI) intervention effects, for N9 and UPG relative to control. Exposure to N9 gel and UPG generated a common "harm signal" that included transcriptional up-regulation of inflammatory genes chemokine (C-C motif) ligand 20 (macrophage inflammatory factor-3alpha) and interleukin 8 in the cervix, decreased protein concentrations of secretory leukocyte protease inhibitor, and transcriptional up-regulation of inflammatory mediators glycodelin-A and osteopontin in the endometrium. These results need to be replicated with a larger sample, but underscore the need to consider the effects of microbicide agents and gel excipients on the upper female reproductive tract in studies of vaginal microbicides.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.