1
|
Martell K, McIntyre JB, Abedin T, Kornaga EN, Chan AMY, Enwere E, Köbel M, Dean ML, Phan T, Ghatage P, Lees-Miller SP, Doll CM. Prevalence and Prognostic Significance of PIK3CA Mutation and CNV Status and Phosphorylated AKT Expression in Patients With Cervical Cancer Treated With Primary Surgery. Int J Gynecol Pathol 2024; 43:158-170. [PMID: 37668363 DOI: 10.1097/pgp.0000000000000978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Currently, there are limited and conflicting reports on the prognostic utility of PIK3CA and associated pathway markers for cervical cancers treated with primary surgical management. Moreover, current studies are lacking complete characterization of adjuvant treatment with RT and/or chemotherapy. We aimed to document the prevalence, clinicopathologic, adjuvant treatment details, and prognostic value of PI3K/AKT pathway mutations and copy number variation and phosphorylated AKT status in patients with cervical cancers treated with primary surgery. A clinicopathologic review was performed on a retrospective cohort of 185 patients with cervical cancer, treated with primary surgery at a single tertiary institution. Next-generation sequencing and digital PCR was used to determine PI3K/AKT pathway mutational status and PIK3CA copy number variation, respectively, and fluorescent immunohistochemistry measured phosphorylated AKT expression. In all, 179 of 185 (96.8%) of tumors were successfully sequenced; 48 (26.8%) were positive for PI3K/AKT pathway mutations-the majority (n=37, 77.1%) PIK3CA mutations. PIK3CA mutation was associated with pathologically positive lymph nodes [12 (32%) vs. 22 (16%); P =0.022] and indication for postoperative chemoradiotherapy [17 (45.9%) vs. 32 (22.5%); P =0.004]. On multivariable analysis, PIK3CA status was not associated with overall survival ( P =0.103) or progression-free survival ( P =0.240) at 5 yrs, nor was PIK3CA copy number variation status. phosphorylated AKT ≤ median significantly predicted for progression-free survival [multivariable hazard ratio 0.39 (0.17-0.89; P =0.025)] but not overall survival ( P =0.087). The correlation of PIK3CA with pathologic positive lymph node status yet lack of association with survival outcomes may be due to the use of adjuvant postoperative therapy. PIK3CA assessment before radical hysterectomy may help identify patients with a higher risk of node-positive disease.
Collapse
|
2
|
Sirohi VK, Medrano TI, Kannan A, Bagchi IC, Cooke PS. Uterine-specific Ezh2 deletion enhances stromal cell senescence and impairs placentation, resulting in pregnancy loss. iScience 2023; 26:107028. [PMID: 37360688 PMCID: PMC10285549 DOI: 10.1016/j.isci.2023.107028] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/10/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Maternal uterine remodeling facilitates embryo implantation, stromal cell decidualization and placentation, and perturbation of these processes may cause pregnancy loss. Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase that epigenetically represses gene transcription; loss of uterine EZH2 affects endometrial physiology and induces infertility. We utilized a uterine Ezh2 conditional knockout (cKO) mouse to determine EZH2's role in pregnancy progression. Despite normal fertilization and implantation, embryo resorption occurred mid-gestation in Ezh2cKO mice, accompanied by compromised decidualization and placentation. Western blot analysis revealed Ezh2-deficient stromal cells have reduced amounts of the histone methylation mark H3K27me3, causing upregulation of senescence markers p21 and p16 and indicating that enhanced stromal cell senescence likely impairs decidualization. Placentas from Ezh2cKO dams on gestation day (GD) 12 show architectural defects, including mislocalization of spongiotrophoblasts and reduced vascularization. In summary, uterine Ezh2 loss impairs decidualization, increases decidual senescence, and alters trophoblast differentiation, leading to pregnancy loss.
Collapse
Affiliation(s)
- Vijay K. Sirohi
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Theresa I. Medrano
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Athilakshmi Kannan
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Indrani C. Bagchi
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Paul S. Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| |
Collapse
|
3
|
Avila M, Grinsfelder MO, Pham M, Westin SN. Targeting the PI3K Pathway in Gynecologic Malignancies. Curr Oncol Rep 2022; 24:1669-1676. [PMID: 36401704 PMCID: PMC10862662 DOI: 10.1007/s11912-022-01326-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2022] [Indexed: 11/21/2022]
Abstract
PURPOSE OF REVIEW This review explores the PI3K pathway aberrations common in gynecologic malignancies, the relevant therapeutic targets that have been explored to date particularly given their success in endometrial cancers, and predictive biomarkers of response to therapy. RECENT FINDINGS Landmark trials have been noted involving this pathway, particularly in endometrial cancers. One phase II trial of the potent orally bioavailable mTOR inhibitor, everolimus, in combination with letrozole demonstrated an unprecedented clinical benefit rate (CBR) of 40% and high objective response rate (RR) of 32% in hormone agnostic endometrial cancers. This was followed by GOG 3007 that compared everolimus and letrozole to hormonal therapy yielding similar response rates but double progression-free survival rates. The phosphoinositide 3-kinase (PI3K) signaling pathway is implicated in tumorigenesis given its regulation over cell growth, cellular trafficking, and angiogenesis. In gynecologic malignancies, alterations in PI3K signaling are common. Therefore, developing modulators of the PI3K pathway and identifying molecular markers to predict response are of great interest for these cancer types.
Collapse
Affiliation(s)
- Monica Avila
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Herman Pressler Dr. CPB 6.3279, Houston, TX, 77030, USA
| | - Michaela Onstad Grinsfelder
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Herman Pressler Dr. CPB 6.3279, Houston, TX, 77030, USA
| | - Melissa Pham
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Herman Pressler Dr. CPB 6.3279, Houston, TX, 77030, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Herman Pressler Dr. CPB 6.3279, Houston, TX, 77030, USA.
| |
Collapse
|
4
|
Mesa AM, Mao J, Medrano TI, Bivens NJ, Jurkevich A, Tuteja G, Cooke PS, Rosenfeld CS. Spatial Transcriptomics analysis of uterine gene expression in enhancer of Zeste homolog 2 (Ezh2) conditional knockout mice. Biol Reprod 2021; 105:1126-1139. [PMID: 34344022 DOI: 10.1093/biolre/ioab147] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/12/2021] [Indexed: 12/16/2022] Open
Abstract
Histone proteins undergo various modifications that alter chromatin structure, including addition of methyl groups. Enhancer of homolog 2 (EZH2), is a histone methyltransferase that methylates lysine residue 27, and thereby, suppresses gene expression. EZH2 plays integral role in the uterus and other reproductive organs. We have previously shown that conditional deletion of uterine EZH2 results in increased proliferation of luminal and glandular epithelial cells, and RNAseq analyses reveal several uterine transcriptomic changes in Ezh2 conditional (c) knockout (KO) mice that can affect estrogen signaling pathways. To pinpoint the origin of such gene expression changes, we used the recently developed spatial transcriptomics (ST) method with the hypotheses that Ezh2cKO mice would predominantly demonstrate changes in epithelial cells and/or ablation of this gene would disrupt normal epithelial/stromal gene expression patterns. Uteri were collected from ovariectomized adult WT and Ezh2cKO mice and analyzed by ST. Asb4, Cxcl14, Dio2, and Igfbp5 were increased, Sult1d1, Mt3, and Lcn2 were reduced in Ezh2cKO uterine epithelium vs. WT epithelium. For Ezh2cKO uterine stroma, differentially expressed key hub genes included Cald1, Fbln1, Myh11, Acta2, and Tagln. Conditional loss of uterine Ezh2 also appears to shift the balance of gene expression profiles in epithelial vs. stromal tissue toward uterine epithelial cell and gland development and proliferation, consistent with uterine gland hyperplasia in these mice. Current findings provide further insight into how EZH2 may selectively affect uterine epithelial and stromal compartments. Additionally, these transcriptome data might provide the mechanistic understanding and valuable biomarkers for human endometrial disorders with epigenetic underpinnings.
Collapse
Affiliation(s)
- Ana M Mesa
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA.,Grupo de Investigación en Agrociencias, Biodiversidad y Territorio - GAMMA, Facultad de Ciencias Agrarias, Universidad de Antioquia UdeA, Calle 70 N° 52-21, Medellín, Colombia
| | - Jiude Mao
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.,Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Theresa I Medrano
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Nathan J Bivens
- Genomics Technology, University of Missouri, Columbia, MO 65211, USA
| | - Alexander Jurkevich
- Advanced Light Microscopy Core Facility, University of Missouri, Columbia, MO 65211, USA
| | - Geetu Tuteja
- Genetics, Development and Cell Biology, Iowa State University, Ames, IA 50011, USA
| | - Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Cheryl S Rosenfeld
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.,Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA.,Data Science and Informatics Institute, University of Missouri, Columbia; MO 65211, USA.,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, 65211, USA
| |
Collapse
|
5
|
Nanjappa MK, Mesa AM, Medrano TI, Jefferson WN, DeMayo FJ, Williams CJ, Lydon JP, Levin ER, Cooke PS. The histone methyltransferase EZH2 is required for normal uterine development and function in mice†. Biol Reprod 2020; 101:306-317. [PMID: 31201420 DOI: 10.1093/biolre/ioz097] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 04/26/2019] [Accepted: 06/06/2019] [Indexed: 01/04/2023] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a rate-limiting catalytic subunit of a histone methyltransferase, polycomb repressive complex, which silences gene activity through the repressive histone mark H3K27me3. EZH2 is critical for epigenetic effects of early estrogen treatment, and may be involved in uterine development and pathologies. We investigated EZH2 expression, regulation, and its role in uterine development/function. Uterine epithelial EZH2 expression was associated with proliferation and was high neonatally then declined by weaning. Pre-weaning uterine EZH2 expression was comparable in wild-type and estrogen receptor 1 knockout mice, showing neonatal EZH2 expression is ESR1 independent. Epithelial EZH2 was upregulated by 17β-estradiol (E2) and inhibited by progesterone in adult uteri from ovariectomized mice. To investigate the uterine role of EZH2, we developed a EZH2 conditional knockout (Ezh2cKO) mouse using a cre recombinase driven by the progesterone receptor (Pgr) promoter that produced Ezh2cKO mice lacking EZH2 in Pgr-expressing tissues (e.g. uterus, mammary glands). In Ezh2cKO uteri, EZH2 was deleted neonatally. These uteri had reduced H3K27me3, were larger than WT, and showed adult cystic endometrial hyperplasia. Ovary-independent uterine epithelial proliferation and increased numbers of highly proliferative uterine glands were seen in adult Ezh2cKO mice. Female Ezh2cKO mice were initially subfertile, and then became infertile by 9 months. Mammary gland development in Ezh2cKO mice was inhibited. In summary, uterine EZH2 expression is developmentally and hormonally regulated, and its loss causes aberrant uterine epithelial proliferation, uterine hypertrophy, and cystic endometrial hyperplasia, indicating a critical role in uterine development and function.
Collapse
Affiliation(s)
- Manjunatha K Nanjappa
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Ana M Mesa
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Theresa I Medrano
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Wendy N Jefferson
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Carmen J Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Ellis R Levin
- Division of Endocrinology, Department of Medicine, University of California-Irvine, Irvine, California, USA.,Department of Veterans Affairs Medical Center, Long Beach, Long Beach, California, USA
| | - Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
6
|
Martell K, McIntyre JB, Kornaga EN, Chan AMY, Phan T, Köbel M, Enwere EK, Dean ML, Ghatage P, Lees-Miller SP, Doll CM. PIK3CA mutation and CNV status and post-chemoradiotherapy survival in patients with cervical cancer. Gynecol Oncol 2020; 158:776-784. [PMID: 32653099 DOI: 10.1016/j.ygyno.2020.06.506] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/25/2020] [Indexed: 02/05/2023]
Abstract
PURPOSE This study aimed to describe the prognostic value of PI3K/AKT pathway mutations in a large cohort of patients with cervical cancer. EXPERIMENTAL DESIGN Patients with pre-treatment archival specimens, diagnosed with FIGO stages IB-IVA cervical cancer between 1998 and 2014 and treated with radical, curative intent chemoradiotherapy (CRT) at a single center were identified. Mutational status was determined by next generation sequencing and PIK3CA copy number (CNV) was assessed by digital PCR. RESULTS 190 patients with available pre-treatment tumor specimens were identified. Median OS and PFS were 57.4 and 46.0 months, respectively. A total of 161 tumors were successfully sequenced; 60 (37.3%) had PI3K/AKT pathway mutations, with 50 (30.1%) having PIK3CA hotspot mutations. PIK3CA CNV gain was noted in 79 (59.2%) of the 154 successfully analyzed. On univariate analysis, PIK3CA mutation was associated with poor OS (HR 1.73; 95% CI: 1.03-2.92; p = .037) but not PFS (HR 1.38; 0.84-2.28; p = .204). Absence of any PI3K/AKT pathway mutation was associated with improved OS (HR 1.68; 1.01-2.81; p = .046) but not PFS (HR 1.50; 0.93-2.43; p = .202). Associations were not maintained when adjusting for clinical factors. On univariate analysis, PIK3CA mutation positive, CNV normal tumors were associated with poorer OS (HR 2.55; 1.18-5.50; p = .017) and trend to worse PFS (HR 1.87; 0.90-3.83; p = .094) when compared to those with CNV gain and wildtype PIK3CA. CONCLUSIONS PI3K/AKT pathway mutations are common in cervical cancer. Consideration of PIK3CA mutational status with CNV status may be important in predicting outcome in cervical cancer patients.
Collapse
Affiliation(s)
- Kevin Martell
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - John B McIntyre
- Precision Oncology Hub, Tom Baker Cancer Centre, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Elizabeth N Kornaga
- Precision Oncology Hub, Tom Baker Cancer Centre, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Angela M Y Chan
- Precision Oncology Hub, Tom Baker Cancer Centre, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Tien Phan
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Emeka K Enwere
- Precision Oncology Hub, Tom Baker Cancer Centre, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Michelle L Dean
- Precision Oncology Hub, Tom Baker Cancer Centre, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Prafull Ghatage
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Susan P Lees-Miller
- Department of Biochemistry and Molecular Biology, Oncology and Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Corinne M Doll
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
7
|
Ogasawara A, Sato S, Hasegawa K. Current and future strategies for treatment of ovarian clear cell carcinoma. J Obstet Gynaecol Res 2020; 46:1678-1689. [PMID: 32578333 DOI: 10.1111/jog.14350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 05/23/2020] [Indexed: 01/04/2023]
Abstract
Ovarian clear cell carcinoma (OCCC) is one of the five histological types of epithelial ovarian cancer (EOC). OCCC comprises 23% of all EOC cases in Japan, whereas the rate of OCCC in North America and Europe is much lower. OCCC is generally categorized as a rare gynecologic malignancy, and there is limited evidence for specific treatment. The clinical basis for treatment of OCCC is mostly based on retrospective studies, many of which were performed in Japan. Until recently, most randomized clinical trials for EOC have included OCCC; therefore, current treatment for OCCC is basically the same as that for other histologic types of EOC. However, the clinical characteristics of OCCC differ from those of high-grade serous carcinoma, particularly for chemosensitivity, and there is a need to develop new treatment for OCCC. The molecular background of OCCC has unique features: tumors are usually negative for p53 mutations and positive for ARID1A and/or PIK3CA mutations, whereas p53 mutations are common in high-grade serous or endometrioid carcinomas. These features may help in development of new treatment for OCCC. In this review, we described the current evidence for treatment of OCCC, including surgery, radiotherapy, chemotherapy, molecular targeted therapy and immunotherapy, and we discuss ongoing clinical trials and preclinical studies of new treatment approaches for OCCC.
Collapse
Affiliation(s)
- Aiko Ogasawara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Sho Sato
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| |
Collapse
|
8
|
Facchinetti F, Appetecchia M, Aragona C, Bevilacqua A, Bezerra Espinola MS, Bizzarri M, D'Anna R, Dewailly D, Diamanti-Kandarakis E, Hernández Marín I, Kamenov ZA, Kandaraki E, Laganà AS, Monastra G, Montanino Oliva M, Nestler JE, Orio F, Ozay AC, Papalou O, Pkhaladze L, Porcaro G, Prapas N, Soulage CO, Stringaro A, Wdowiak A, Unfer V. Experts' opinion on inositols in treating polycystic ovary syndrome and non-insulin dependent diabetes mellitus: a further help for human reproduction and beyond. Expert Opin Drug Metab Toxicol 2020; 16:255-274. [PMID: 32129111 DOI: 10.1080/17425255.2020.1737675] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/28/2020] [Indexed: 01/05/2023]
Abstract
Introduction: This Experts' opinion provides an updated scientific support to gynecologists, obstetricians, endocrinologists, nutritionists, neurologists and general practitioners on the use of Inositols in the therapy of Polycystic Ovary Syndrome (PCOS) and non-insulin dependent (type 2) diabetes mellitus (NIDDM).Areas covered: This paper summarizes the physiology of Myo-Inositol (MI) and D-Chiro-Inositol (DCI), two important molecules present in human organisms, and their therapeutic role, also for treating infertility. Some deep differences between the physiological functions of MI and DCI, as well as their safety and intestinal absorption are discussed. Updates include new evidence on the efficacy exerted in PCOS by the 40:1 MI/DCI ratio, and the innovative approach based on alpha-lactalbumin to overcome the decreased therapeutic efficacy of Inositols in some patients.Expert opinion: The evidence suggests that MI, alone or with DCI in the 40:1 ratio, offers a promising treatment for PCOS and NIDDM. However, additional studies need to evaluate some still unresolved issues, such as the best MI/DCI ratio for treating NIDDM, the potential cost-effectiveness of reduced gonadotropins administration in IVF due to MI treatment, or the benefit of MI supplementation in ovulation induction with clomiphene citrate in PCOS patients.
Collapse
Affiliation(s)
- Fabio Facchinetti
- Department of Obstetrics and Gynecology and Pediatrics, University of Modena and Reggio Emilia, Modena, Italy
| | - Marialuisa Appetecchia
- Oncological Endocrinology Unit, Regina Elena National Cancer Institute - IRCCS, Rome, Italy
| | - Cesare Aragona
- Systems Biology Group Lab, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Arturo Bevilacqua
- Department of Dynamic and Clinical Psychology, Sapienza University of Rome, Rome, Italy
| | | | - Mariano Bizzarri
- Systems Biology Group Lab, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Rosario D'Anna
- Unit of Gynecology and Obstetrics, Department of Human Pathology in Adulthood and Childhood "G. Barresi", University of Messina, Messina, Italy
| | - Didier Dewailly
- Faculty of Medicine, University of Lille, Lille, France
- INSERM, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, Lille, France
| | | | - Imelda Hernández Marín
- Human Reproduction Department, Hospital Juárez de México, México City Mexico
- Facultad de Medicina, Universidad Nacional Autónoma De México (UNAM), México City, México
| | - Zdravko A Kamenov
- Department of Internal Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Eleni Kandaraki
- Department of Endocrinology & Diabetes, HYGEIA Hospital, Marousi, Athens, Greece
| | - Antonio Simone Laganà
- Department of Obstetrics and Gynecology, "Filippo Del Ponte" Hospital, University of Insubria, Varese, Italy
| | - Giovanni Monastra
- Systems Biology Group Lab, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - John E Nestler
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Francesco Orio
- Department of Endocrinology, "Parthenope" University of Naples, Italy
| | - Ali Cenk Ozay
- Faculty of Medicine, Department of Obstetrics and Gynecology, Near East University, Nicosia Cyprus
- Near East University, Research Center of Experimental Health Sciences, Nicosia, Cyprus
| | - Olga Papalou
- Department of Endocrinology & Diabetes, HYGEIA Hospital, Marousi, Athens, Greece
| | - Lali Pkhaladze
- Department of Gynecological Endocrinology, Ioseb Zhordania Institute of Reproductology, Tbilisi, Georgia
| | | | - Nikos Prapas
- 3rd Department of OB-GYNAE, Aristotle University of Thessaloniki, Thessaloniki Greece
- IVF Laboratory, IAKENTRO Fertility Centre, Thessaloniki, Greece
| | | | - Annarita Stringaro
- National Center for Drug Research and Evaluation - Italian National Institute of Health, Rome, Italy
| | - Artur Wdowiak
- Diagnostic Techniques Unit, Medical University of Lublin, Poland
| | - Vittorio Unfer
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
9
|
Mesa AM, Mao J, Nanjappa MK, Medrano TI, Tevosian S, Yu F, Kinkade J, Lyu Z, Liu Y, Joshi T, Wang D, Rosenfeld CS, Cooke PS. Mice lacking uterine enhancer of zeste homolog 2 have transcriptomic changes associated with uterine epithelial proliferation. Physiol Genomics 2020; 52:81-95. [PMID: 31841397 PMCID: PMC7052568 DOI: 10.1152/physiolgenomics.00098.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/21/2019] [Accepted: 12/09/2019] [Indexed: 01/16/2023] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase that suppresses gene expression. Previously, we developed a conditional null model where EZH2 is knocked out in uterus. Deletion of uterine EZH2 increased proliferation of luminal and glandular epithelial cells. Herein, we used RNA-Seq in wild-type (WT) and EZH2 conditional knockout (Ezh2cKO) uteri to obtain mechanistic insights into the gene expression changes that underpin the pathogenesis observed in these mice. Ovariectomized adult Ezh2cKO mice were treated with vehicle (V) or 17β-estradiol (E2; 1 ng/g). Uteri were collected at postnatal day (PND) 75 for RNA-Seq or immunostaining for epithelial proliferation. Weighted gene coexpression network analysis was used to link uterine gene expression patterns and epithelial proliferation. In V-treated mice, 88 transcripts were differentially expressed (DEG) in Ezh2cKO mice, and Bmp5, Crabp2, Lgr5, and Sprr2f were upregulated. E2 treatment resulted in 40 DEG with Krt5, Krt15, Olig3, Crabp1, and Serpinb7 upregulated in Ezh2cKO compared with control mice. Transcript analysis relative to proliferation rates revealed two module eigengenes correlated with epithelial proliferation in WT V vs. Ezh2cKO V and WT E2 vs. Ezh2cKO E2 mice, with a positive relationship in the former and inverse in the latter. Notably, the ESR1, Wnt, and Hippo signaling pathways were among those functionally enriched in Ezh2cKO females. Current results reveal unique gene expression patterns in Ezh2cKO uterus and provide insight into how loss of this critical epigenetic regulator assumingly contributes to uterine abnormalities.
Collapse
Affiliation(s)
- Ana M Mesa
- Department of Physiological Sciences, University of Florida, Gainesville, Florida
- Grupo de Investigación en Génetica, Mejoramiento y Modelación Animal-GaMMA, Universidad de Antioquia, Medellín, Colombia
| | - Jiude Mao
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Biomedical Sciences, University of Missouri, Columbia, Missouri
| | | | - Theresa I Medrano
- Department of Physiological Sciences, University of Florida, Gainesville, Florida
| | - Sergei Tevosian
- Department of Physiological Sciences, University of Florida, Gainesville, Florida
| | - Fahong Yu
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Missouri
| | - Jessica Kinkade
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Zhen Lyu
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri
| | - Yang Liu
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Informatics Institute, University of Missouri, Columbia; Missouri
| | - Trupti Joshi
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri
- Informatics Institute, University of Missouri, Columbia; Missouri
- Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, Missouri
| | - Duolin Wang
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri
| | - Cheryl S Rosenfeld
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Biomedical Sciences, University of Missouri, Columbia, Missouri
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Missouri
- Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, Missouri
| | - Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, Florida
| |
Collapse
|
10
|
Shi X, Wang J, Lei Y, Cong C, Tan D, Zhou X. Research progress on the PI3K/AKT signaling pathway in gynecological cancer (Review). Mol Med Rep 2019; 19:4529-4535. [PMID: 30942405 PMCID: PMC6522820 DOI: 10.3892/mmr.2019.10121] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 02/21/2019] [Indexed: 02/06/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway is involved in the regulation of multiple cellular physiological processes by activating downstream corresponding effector molecules, which serve an important role in the cell cycle, growth and proliferation. This is a common phenomenon; overactivation of the pathway is present in human malignancies and has been implicated in cancer progression, hence one of the important approaches to the treatment of tumors is rational drug design using molecular targets in the PI3K/AKT signaling pathway. In brief, the present review analyzed the effects of the PI3K/AKT signaling pathway on certain gynecological cancer types.
Collapse
Affiliation(s)
- Xiang Shi
- Department of Pathology, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Jingjing Wang
- Department of Pathology, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Yu Lei
- Department of Pathology, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Caofan Cong
- Department of Pathology, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Dailin Tan
- Department of Clinical Laboratory, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Xianrong Zhou
- Department of Pathology, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| |
Collapse
|
11
|
Cernaj IE. Simultaneous dual targeting of Par-4 and G6PD: a promising new approach in cancer therapy? Quintessence of a literature review on survival requirements of tumor cells. Cancer Cell Int 2016; 16:87. [PMID: 27872579 PMCID: PMC5111342 DOI: 10.1186/s12935-016-0363-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/07/2016] [Indexed: 11/10/2022] Open
Abstract
The aim of this hypothesis is to propose a new approach in targeted therapy of cancer: The simultaneous, dual targeting of two single molecules, Par-4 and G6PD, rather than inhibition of full-length signaling pathways. RATIONALE Targeted inhibition of especially two survival signaling pathways (PI3K/AKT/mTOR and MAPK/ERK) is frequently tried, however, a major breakthrough has not yet been reported. Inhibition of complete pathways naturally goes along with a variety of dose-limiting side effects thus contributing to poor efficacy of the administered drugs. This essay offers a synopsis of relevant studies to support the above mentioned idea-targeting of two single molecules which either are crucial for tumor growth and cancer-cell-survival: on one side, Par-4-activation selectively triggers apoptosis of tumor cells thus reversing their characteristic feature-immortality. On the other side inhibition of G6PD breaks the energy supply of tumor cells, weakens their defence against oxidative stress and thereby enhances the sensitivity of tumor cells to oxidative agents (e.g. chemotherapy). Advantage of the proposed dual Par-4/G6PD-therapy is good tolerability and-especially when administered along with conventional therapy-less frequent emergence of resistance.
Collapse
|
12
|
Yang SD, Jang SS, Han JA, Park HS, Kim JI. Estimation of Prognostic Marker Genes by Public Microarray Data in Patients with Ovarian Serous Cystadenocarcinoma. Yonsei Med J 2016; 57:872-8. [PMID: 27189279 PMCID: PMC4951462 DOI: 10.3349/ymj.2016.57.4.872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/03/2015] [Accepted: 10/08/2015] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Lymphatic invasion (LI) is regarded as a predictor of the aggressiveness of ovarian cancer (OC). However, LI is not always the major determinant of long-term patient survival. To establish proper diagnosis and treatment for OC, we analyzed differentially expressed genes (DEGs) for patients with serous epithelial OC, with or without LI, who did or did not survive for 5 years. MATERIALS AND METHODS Gene expression data from 63 patients with OC and LI, and 35 patients with OC but without LI, were investigated using an Affymetrix Human Genome U133 Array and analyzed using The Cancer Genome Atlas (TCGA) database. Among these 98 patients, 16 survived for 5 years or more. DEGs were identified using the Bioconductor R package, and their functions were analyzed using the DAVID web tool. RESULTS We found 55 significant DEGs (p<0.01) from the patients with LI and 20 highly significant DEGs (p<0.001) from those without it. Pathway analysis showed that DEGs associated with carbohydrate metabolism or with renal cell carcinoma pathways were enriched in the patients with and without LI, respectively. Using the top five prognostic marker genes, we generated survival scores that could be used to predict the 5-year survival of patients with OC without LI. CONCLUSION The DEGs identified in this study could be used to elucidate the mechanism of tumor progression and to guide the prognosis and treatment of patients with serous OC but without LI.
Collapse
Affiliation(s)
- San Duk Yang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Se Song Jang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Jeong A Han
- Department of Biochemistry and Molecular Biology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Hyun Seok Park
- Center for Convergence Research of Advanced Technologies, Ewha Womans University, Seoul, Korea
| | - Jong Il Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea.
| |
Collapse
|
13
|
Lauretta R, Lanzolla G, Vici P, Mariani L, Moretti C, Appetecchia M. Insulin-Sensitizers, Polycystic Ovary Syndrome and Gynaecological Cancer Risk. Int J Endocrinol 2016; 2016:8671762. [PMID: 27725832 PMCID: PMC5048026 DOI: 10.1155/2016/8671762] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/12/2016] [Accepted: 08/08/2016] [Indexed: 12/28/2022] Open
Abstract
Preclinical, early phase clinical trials and epidemiological evidence support the potential role of insulin-sensitizers in cancer prevention and treatment. Insulin-sensitizers improve the metabolic and hormonal profile in PCOS patients and may also act as anticancer agents, especially in cancers associated with hyperinsulinemia and oestrogen dependent cancers. Several lines of evidence support the protection against cancer exerted by dietary inositol, in particular inositol hexaphosphate. Metformin, thiazolidinediones, and myoinositol postreceptor signaling may exhibit direct inhibitory effects on cancer cell growth. AMPK, the main molecular target of metformin, is emerging as a target for cancer prevention and treatment. PCOS may be correlated to an increased risk for developing ovarian and endometrial cancer (up to threefold). Several studies have demonstrated an increase in mortality rate from ovarian cancer among overweight/obese PCOS women compared with normal weight women. Long-term use of metformin has been associated with lower rates of ovarian cancer. Considering the evidence supporting a higher risk of gynaecological cancer in PCOS women, we discuss the potential use of insulin-sensitizers as a potential tool for chemoprevention, hypothesizing a possible rationale through which insulin-sensitizers may inhibit tumourigenesis.
Collapse
Affiliation(s)
- Rosa Lauretta
- Unit of Endocrinology, Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Lanzolla
- Unit of Endocrinology, Department of Systems' Medicine, University of Rome Tor Vergata, Section of Reproductive Endocrinology, Fatebenefratelli Hospital “San Giovanni Calibita” Rome, Italy
| | - Patrizia Vici
- Division of Medical Oncology B, Regina Elena National Cancer Institute, Rome, Italy
| | - Luciano Mariani
- Department of Gynaecologic Oncology, HPV-Unit, Regina Elena National Cancer Institute, Rome, Italy
| | - Costanzo Moretti
- Unit of Endocrinology, Department of Systems' Medicine, University of Rome Tor Vergata, Section of Reproductive Endocrinology, Fatebenefratelli Hospital “San Giovanni Calibita” Rome, Italy
| | - Marialuisa Appetecchia
- Unit of Endocrinology, Regina Elena National Cancer Institute, Rome, Italy
- *Marialuisa Appetecchia:
| |
Collapse
|
14
|
Lu D, Qian J, Li W, Feng Q, Pan S, Zhang S. β-hydroxyisovaleryl-shikonin induces human cervical cancer cell apoptosis via PI3K/AKT/mTOR signaling. Oncol Lett 2015; 10:3434-3442. [PMID: 26788147 PMCID: PMC4665374 DOI: 10.3892/ol.2015.3769] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 06/22/2015] [Indexed: 12/31/2022] Open
Abstract
The present study aimed to investigate the inhibitory ability of β-hydroxyisovaleryl-shikonin (β-HIVS) on the proliferation of human cervical cancer HeLa cells and to identify the mechanism of this effect. The HeLa cells were treated with β-HIVS and the inhibition of cell growth was detected by an MTT assay. Flow cytometry was performed to analyze the apoptosis rate and cell cycle distribution of HeLa cells. Reverse transcription-polymerase chain reaction and western blot analysis were used to examine the expression of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway proteins. The results revealed that β-HIVS inhibited HeLa cell proliferation in a dose- and time-dependent manner. With the administration of increasing concentrations of β-HIVS, the apoptotic rate of HeLa cells was also increased. The cell cycle was slightly arrested at the S phase, with ~6% of cells in this phase, subsequent to treatment with 10 µM β-HIVS. In addition, β-HIVS markedly reduced the expression levels of PI3K, AKT, mTOR and 70-kDa ribosomal protein S6 kinase in HeLa cells. β-HIVS promoted cervical cancer cell apoptosis by inhibiting the PI3K/AKT/mTOR signaling pathway and suppressing downstream gene expression. The present study is expected to lead to the development of molecular targeted therapy for this signaling pathway as a novel method of cervical cancer treatment.
Collapse
Affiliation(s)
- Dan Lu
- Department of Gynecology & Obstetrics, College of Clinical Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Jing Qian
- Department of Gynecology & Obstetrics, College of Clinical Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Wei Li
- Department of Gynecology & Obstetrics, College of Clinical Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Qianqian Feng
- Department of Gynecology & Obstetrics, College of Clinical Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Shu Pan
- Department of Gynecology & Obstetrics, College of Clinical Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Siquan Zhang
- Department of Gynecology & Obstetrics, College of Clinical Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| |
Collapse
|
15
|
Yamashita Y. Ovarian cancer: new developments in clear cell carcinoma and hopes for targeted therapy. Jpn J Clin Oncol 2015; 45:405-7. [PMID: 25583423 PMCID: PMC4412138 DOI: 10.1093/jjco/hyu221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 12/19/2014] [Indexed: 12/30/2022] Open
Abstract
Until recently, ovarian clear cell carcinoma was recognized by its unique morphology and unfavorable patient outcome primarily due to tumor chemoresistance. Recently, specific molecular characteristics of ovarian clear cell carcinoma, such as PI3CA mutation, ARID1a mutation and MET amplification, have been elucidated. In addition, an association between endometriosis and the tumor has also been a focus of research in recent years. The aim of this review is to discuss the specificity and importance of molecular changes and various intriguing points that are not solved until today. Finally, future aspects, including hopes for the development of novel therapies, are discussed.
Collapse
Affiliation(s)
- Yoriko Yamashita
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|