1
|
Koinuma Y, Mitsuishi Y, Winardi W, Hidayat M, Wirawan A, Hayakawa D, Kanamori K, Matsumoto N, Hayashi T, Shimada N, Tajima K, Takamochi K, Takahashi F, Suzuki K, Takahashi K. Anti-Müllerian hormone type II receptor protein expression in non-small cell lung cancer and the effect of AMH/AMHR2 signaling on cancer cell proliferation. Thorac Cancer 2024; 15:2090-2099. [PMID: 39230026 PMCID: PMC11471453 DOI: 10.1111/1759-7714.15309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide despite advances in cancer therapeutics. In several gynecological cancers, anti-Müllerian hormone receptor type 2 (AMHR2) mediates AMH-induced growth inhibition and is expressed at high levels. Furthermore, 5%-8% of NSCLCs exhibit high AMHR2 expression, suggesting that AMH may inhibit the progression of some lung cancers. However, the clinical relevance of AMHR2 expression and its role in lung cancer is not fully clarified. METHODS Immunostaining was performed on 79 surgical specimens of NSCLC. The Cancer Genome Atlas RNA-seq data for lung adenocarcinoma were analyzed, and gene ontology and gene set enrichment analyses were performed. In cellular experiments, AMHR2-overexpressing NSCLC cell lines were established, and the role of the AMH-AMHR2 pathway in cell proliferation with recombinant human AMH protein treatment was examined. RESULTS A total of 13 cases (16.5%) were positive for immunostaining in lung adenocarcinoma tissues; no positive signals were detected in lung squamous carcinoma tissues. Gene expression variation analysis using The Cancer Genome Atlas data showed that the expression of genes related to the cell cycle was downregulated in the AMHR2-high group. Cellular experiments showed that activation of the AMH-AMHR2 pathway suppressed cell proliferation. CONCLUSION In lung adenocarcinoma tissues with high expression of AMHR2, activation of the AMH-AMHR2 pathway may suppress cell proliferation.
Collapse
Affiliation(s)
- Yoshika Koinuma
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| | - Yoichiro Mitsuishi
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| | - Wira Winardi
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| | - Moulid Hidayat
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| | - Aditya Wirawan
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| | - Daisuke Hayakawa
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| | - Koichiro Kanamori
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| | - Naohisa Matsumoto
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| | - Takuo Hayashi
- Department of Human PathologyJuntendo University, Graduate School of MedicineTokyoJapan
| | - Naoko Shimada
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| | - Ken Tajima
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| | - Kazuya Takamochi
- Department of General Thoracic SurgeryJuntendo University, Graduate School of MedicineTokyoJapan
| | - Fumiyuki Takahashi
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| | - Kenji Suzuki
- Department of General Thoracic SurgeryJuntendo University, Graduate School of MedicineTokyoJapan
| | - Kazuhisa Takahashi
- Department of Respiratory MedicineJuntendo University, Graduate School of MedicineTokyoJapan
- Research Institute for Diseases of Old AgesJuntendo University, Graduate School of MedicineTokyoJapan
| |
Collapse
|
2
|
Gowkielewicz M, Lipka A, Zdanowski W, Waśniewski T, Majewska M, Carlberg C. Anti-Müllerian hormone: biology and role in endocrinology and cancers. Front Endocrinol (Lausanne) 2024; 15:1468364. [PMID: 39351532 PMCID: PMC11439669 DOI: 10.3389/fendo.2024.1468364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Anti-Müllerian hormone (AMH) is a peptide belonging to the transforming growth factor beta superfamily and acts exclusively through its receptor type 2 (AMHR2). From the 8th week of pregnancy, AMH is produced by Sertoli cells, and from the 23rd week of gestation, it is produced by granulosa cells of the ovary. AMH plays a critical role in regulating gonadotropin secretion, ovarian tissue responsiveness to pituitary hormones, and the pathogenesis of polycystic ovarian syndrome. It inhibits the transition from primordial to primary follicles and is considered the best marker of ovarian reserve. Therefore, measuring AMH concentration of the hormone is valuable in managing assisted reproductive technologies. AMH was initially discovered through its role in the degeneration of Müllerian ducts in male fetuses. However, due to its ability to inhibit the cell cycle and induce apoptosis, it has also garnered interest in oncology. For example, antibodies targeting AMHR2 are being investigated for their potential in diagnosing and treating various cancers. Additionally, AMH is present in motor neurons and functions as a protective and growth factor. Consequently, it is involved in learning and memory processes and may support the treatment of Alzheimer's disease. This review aims to provide a comprehensive overview of the biology of AMH and its role in both endocrinology and oncology.
Collapse
Affiliation(s)
- Marek Gowkielewicz
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Aleksandra Lipka
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Wojciech Zdanowski
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Tomasz Waśniewski
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Marta Majewska
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
3
|
Mizuno R, Yamaguchi R, Matsuura K, Ishigami A, Sakumoto R, Sawai K, Koyama K, Okubo M, Souma K, Hirayama H. Expression and localization of anti-Müllerian hormone and its receptors in bovine corpus luteum. Theriogenology 2024; 226:228-235. [PMID: 38924892 DOI: 10.1016/j.theriogenology.2024.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/16/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Although anti-Müllerian hormone (AMH) is involved in the regulation of granulosa cell function in female animals, its role in tissues other than ovarian follicles remains poorly understood. It has also been suggested that cows with high circulating AMH concentrations have increased fertility; however, the mechanism has not been elucidated. This study was conducted to identify the presence of the AMH-signaling system and its target cells in the bovine corpus luteum formed from an ovulated follicle. Immunoblotting revealed that the proteolytically cleaved C-terminal region in AMH (AMHC), a biologically active peptide, was present in trace amounts in the early corpus luteum and significantly increased during the mid to regressed stages. AMHC and cleaved N-terminal region (AMHN) in AMH generate a noncovalent isoform that improves the activity of AMH signaling. An immunohistochemical analysis revealed that AMHC, AMHN, and type II AMH receptor (AMHR2) were localized to luteal cells during the entire estrous cycle. AMH in the corpus luteum seemed to be newly synthesized since AMH expression was detected. These findings suggest that AMH signaling is involved in the regulation of luteal cell function through an autocrine and post-translational processing mechanism. The level of AMHR2 and mRNA expression of AMHR2 and type I AMH receptors (activin-like kinase 2, 3, and 6) were highest in the mid stage. Thus, AMH signaling in the corpus luteum may also be regulated by changes in the receptor levels. Since the transforming growth factor-beta superfamily, to which AMH belongs, is a multifunctional polypeptide growth factor, further studies are needed to evaluate whether AMH signaling has a role in facilitating or inhibiting luteal cell functions.
Collapse
Affiliation(s)
- Riuru Mizuno
- Department of Bioproduction, Graduate School of Bioindustry, Tokyo University of Agriculture, Abashiri, Hokkaido, 099-2493, Japan
| | - Rin Yamaguchi
- Department of Bioproduction, Faculty of Bioindustry, Tokyo University of Agriculture, Abashiri, Hokkaido, 099-2493, Japan
| | - Kaoru Matsuura
- Department of Bioproduction, Faculty of Bioindustry, Tokyo University of Agriculture, Abashiri, Hokkaido, 099-2493, Japan
| | - Ayaha Ishigami
- Department of Bioproduction, Graduate School of Bioindustry, Tokyo University of Agriculture, Abashiri, Hokkaido, 099-2493, Japan
| | - Ryosuke Sakumoto
- Division of Advanced Feeding Technology Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Ibaraki, 305-0901, Japan
| | - Ken Sawai
- The United Graduate School of Agricultural Sciences, Iwate University, Iwate, 020-8550, Japan
| | - Keisuke Koyama
- Laboratory of Theriogenology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, 598-8531, Japan
| | - Michiko Okubo
- Department of Bioproduction, Faculty of Bioindustry, Tokyo University of Agriculture, Abashiri, Hokkaido, 099-2493, Japan
| | - Kousaku Souma
- Department of Bioproduction, Graduate School of Bioindustry, Tokyo University of Agriculture, Abashiri, Hokkaido, 099-2493, Japan; Department of Bioproduction, Faculty of Bioindustry, Tokyo University of Agriculture, Abashiri, Hokkaido, 099-2493, Japan
| | - Hiroki Hirayama
- Department of Bioproduction, Graduate School of Bioindustry, Tokyo University of Agriculture, Abashiri, Hokkaido, 099-2493, Japan; Department of Bioproduction, Faculty of Bioindustry, Tokyo University of Agriculture, Abashiri, Hokkaido, 099-2493, Japan.
| |
Collapse
|
4
|
Orozco-Castaño C, Mejia-Garcia A, Zambrano Y, Combita AL, Parra-Medina R, Bonilla DA, González A, Odriozola A. Construction of an immune gene expression meta signature to assess the prognostic risk of colorectal cancer patients. ADVANCES IN GENETICS 2024; 112:207-254. [PMID: 39396837 DOI: 10.1016/bs.adgen.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Despite recent advancements in colorectal cancer (CRC) treatment, particularly with the introduction of immunotherapy and checkpoint inhibitors, the efficacy of these therapies remains limited to a subset of patients. To address this challenge, our study aimed to develop a prognostic biomarker based on immune-related genes to predict better outcomes in CRC patients and aid in treatment decision-making. We comprehensively analysed immune gene expression signatures associated with CRC prognosis to construct an immune meta-signature with prognostic potential. Utilising data from The Cancer Genome Atlas (TCGA), we employed Cox regression to identify immune-related genes with prognostic significance from multiple studies. Subsequently, we compared the expression levels of immune genes, levels of immune cell infiltration, and various immune-related molecules between high-risk and low-risk patient groups. Functional analysis using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses provided insights into the biological pathways associated with the identified prognostic genes. Finally, we validated our findings using a separate CRC cohort from the Gene Expression Omnibus (GEO). Integration of the prognostic genes revealed significant disparities in survival outcomes. Differential expression analysis identified a set of immune-associated genes, which were further refined using LASSO penalisation and Cox regression. Univariate Cox regression analyses confirmed the autonomy of the gene signature as a prognostic indicator for CRC patient survival. Our risk prediction model effectively stratified CRC patients based on their prognosis, with the high-risk group showing enrichment in pro-oncogenic terms and pathways. Immune infiltration analysis revealed an augmented presence of certain immunosuppressive subsets in the high-risk group. Finally, we validated the performance of our prognostic model by applying the risk score equation to a different CRC patient dataset, confirming its prognostic potential in this new cohort. Overall, our study presents a novel immune-related gene signature with promising implications for predicting cancer progression and prognosis, thereby enabling more personalised management strategies for CRC patients.
Collapse
Affiliation(s)
- Carlos Orozco-Castaño
- Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología (INC), Bogotá, Colombia; Grupo de Apoyo y Seguimiento para la Investigación GASPI, Instituto Nacional de Cancerología (INC), Bogotá, Colombia.
| | - Alejandro Mejia-Garcia
- Department of Human Genetics, McGill University, Montreal, QC, Canada, McGill University, Genome Centre, Montreal, QC, Canada
| | - Yina Zambrano
- Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología (INC), Bogotá, Colombia
| | - Alba Lucia Combita
- Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología (INC), Bogotá, Colombia; Grupo de Apoyo y Seguimiento para la Investigación GASPI, Instituto Nacional de Cancerología (INC), Bogotá, Colombia; Departamento de Microbiología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Rafael Parra-Medina
- Research Institute, Fundación Universitaria de Ciencias de la Salud-FUCS, Bogotá, Colombia; Department of Pathology, Instituto Nacional de Cancerología, Electronic address, Bogotá, Colombia
| | - Diego A Bonilla
- Research Division, Dynamical Business & Science Society - DBSS International SAS, Bogotá, Colombia; Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Adrián Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| |
Collapse
|
5
|
Han WJ, He P. A novel tumor microenvironment-related gene signature with immune features for prognosis of lung squamous cell carcinoma. J Cancer Res Clin Oncol 2023; 149:13137-13154. [PMID: 37479755 DOI: 10.1007/s00432-023-05042-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 06/28/2023] [Indexed: 07/23/2023]
Abstract
PURPOSE Lung squamous cell carcinoma (LUSC) is an aggressive subset of non-small-cell lung cancer (NSCLC). The tumor microenvironment (TME) plays an important role in the development of LUSC. We aim to identify potential therapeutic targets and a TME-related prognostic signature and for LUSC. METHODS TME-related genes were obtained from TCGA-LUSC dataset. LUSC samples were clustered by the non-negative matrix clustering algorithm (NMF). The prognostic signature was constructed through univariate Cox regression, multivariate Cox regression, and the least absolute shrinkage and selection operator (LASSO) analyses. Gene set enrichment analysis (GSEA) was carried out to explore the enrichment pathways. RESULTS This study constructed a prognostic signature which contained 12 genes: HHIPL2, PLK4, SLC6A4, LSM1, TSLP, P4HA1, AMH, CLDN5, NRTN, CDH2, PTGIS, and STX1A. Patients were classified into high-risk and low-risk groups according to the median risk score of this signature. Compared with low-risk group patients, patients in high-risk group patients had poorer overall survival, which demonstrated this signature was an independent prognostic factor. Besides, correlation analysis and GSEA results revealed that genes of this signature were correlated with immune cells and drug response. CONCLUSION Our novel signature based on 12 TME-related genes might be applied as an independent prognostic indicator. Importantly, the signature could be a promising biomarker and accurately predict the prognosis of LUSC patients.
Collapse
Affiliation(s)
- Wan Jia Han
- Beijing Normal University, Beijing, China.
- Sichuan Second Hospital of TCM, Chengdu, China.
| | - Pengzhi He
- Beijing Normal University, Beijing, China
- Sichuan Second Hospital of TCM, Chengdu, China
| |
Collapse
|
6
|
Wits M, Becher C, de Man F, Sanchez-Duffhues G, Goumans MJ. Sex-biased TGFβ signalling in pulmonary arterial hypertension. Cardiovasc Res 2023; 119:2262-2277. [PMID: 37595264 PMCID: PMC10597641 DOI: 10.1093/cvr/cvad129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/21/2023] [Accepted: 07/04/2023] [Indexed: 08/20/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare cardiovascular disorder leading to pulmonary hypertension and, often fatal, right heart failure. Sex differences in PAH are evident, which primarily presents with a female predominance and increased male severity. Disturbed signalling of the transforming growth factor-β (TGFβ) family and gene mutations in the bone morphogenetic protein receptor 2 (BMPR2) are risk factors for PAH development, but how sex-specific cues affect the TGFβ family signalling in PAH remains poorly understood. In this review, we aim to explore the sex bias in PAH by examining sex differences in the TGFβ signalling family through mechanistical and translational evidence. Sex hormones including oestrogens, progestogens, and androgens, can determine the expression of receptors (including BMPR2), ligands, and soluble antagonists within the TGFβ family in a tissue-specific manner. Furthermore, sex-related genetic processes, i.e. Y-chromosome expression and X-chromosome inactivation, can influence the TGFβ signalling family at multiple levels. Given the clinical and mechanistical similarities, we expect that the conclusions arising from this review may apply also to hereditary haemorrhagic telangiectasia (HHT), a rare vascular disorder affecting the TGFβ signalling family pathway. In summary, we anticipate that investigating the TGFβ signalling family in a sex-specific manner will contribute to further understand the underlying processes leading to PAH and likely HHT.
Collapse
Affiliation(s)
- Marius Wits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Clarissa Becher
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Frances de Man
- Department of Pulmonary Medicine, Amsterdam University Medical Center (UMC) (Vrije Universiteit), 1081 HV Amsterdam, The Netherlands
| | - Gonzalo Sanchez-Duffhues
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| |
Collapse
|
7
|
Chauvin M, Meinsohn MC, Dasari S, May P, Iyer S, Nguyen NMP, Oliva E, Lucchini Z, Nagykery N, Kashiwagi A, Mishra R, Maser R, Wells J, Bult CJ, Mitra AK, Donahoe PK, Pépin D. Cancer-associated mesothelial cells are regulated by the anti-Müllerian hormone axis. Cell Rep 2023; 42:112730. [PMID: 37453057 DOI: 10.1016/j.celrep.2023.112730] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/27/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Cancer-associated mesothelial cells (CAMCs) in the tumor microenvironment are thought to promote growth and immune evasion. We find that, in mouse and human ovarian tumors, cancer cells express anti-Müllerian hormone (AMH) while CAMCs express its receptor AMHR2, suggesting a paracrine axis. Factors secreted by cancer cells induce AMHR2 expression during their reprogramming into CAMCs in mouse and human in vitro models. Overexpression of AMHR2 in the Met5a mesothelial cell line is sufficient to induce expression of immunosuppressive cytokines and growth factors that stimulate ovarian cancer cell growth in an AMH-dependent way. Finally, syngeneic cancer cells implanted in transgenic mice with Amhr2-/- CAMCs grow significantly slower than in wild-type hosts. The cytokine profile of Amhr2-/- tumor-bearing mice is altered and their tumors express less immune checkpoint markers programmed-cell-death 1 (PD1) and cytotoxic T lymphocyte-associated protein 4 (CTLA4). Taken together, these data suggest that the AMH/AMHR2 axis plays a critical role in regulating the pro-tumoral function of CAMCs in ovarian cancer.
Collapse
Affiliation(s)
- M Chauvin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - M-C Meinsohn
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - S Dasari
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA
| | - P May
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - S Iyer
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - N M P Nguyen
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - E Oliva
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Z Lucchini
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - N Nagykery
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - A Kashiwagi
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - R Mishra
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - R Maser
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA
| | - J Wells
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA
| | - C J Bult
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA
| | - A K Mitra
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA
| | - Patricia K Donahoe
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - D Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA; Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA.
| |
Collapse
|
8
|
Ruan F, Liu C, Wang Y, Cao X, Tang Z, Xu J, Zeng J, Yin H, Zheng N, Yang C, Zuo Z, He C. Role of RNA m 6A modification in titanium dioxide nanoparticle-induced acute pulmonary injury: An in vitro and in vivo study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 311:119986. [PMID: 36007795 DOI: 10.1016/j.envpol.2022.119986] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 06/15/2023]
Abstract
RNA N6-methyladenosine (m6A) modification regulates the cell stress response and homeostasis, but whether titanium dioxide nanoparticle (nTiO2)-induced acute pulmonary injury is associated with the m6A epitranscriptome and the underlying mechanisms remain unclear. Here, the potential association between m6A modification and the bioeffects of several engineered nanoparticles (nTiO2, nAg, nZnO, nFe2O3, and nCuO) were verified thorough in vitro experiments. nFe2O3, nZnO, and nTiO2 exposure significantly increased the global m6A level in A549 cells. Our study further revealed that nTiO2 can induce m6A-mediated acute pulmonary injury. Mechanistically, nTiO2 exposure promoted methyltransferase-like 3 (METTL3)-mediated m6A signal activation and thus mediated the inflammatory response and IL-8 release through the degeneration of anti-Mullerian hormone (AMH) and Mucin5B (MUC5B) mRNAs in a YTH m6A RNA-binding protein 2 (YTHDF2)-dependent manner. Moreover, nTiO2 exposure stabilized METTL3 protein by the lipid reactive oxygen species (ROS)-activated ERK1/2 pathway. The scavenging of ROS with ferrostatin-1 (Fer-1) alleviates the ERK1/2 activation, m6A upregulation, and the inflammatory response caused by nTiO2 both in vitro and in vivo. In conclusion, our study demonstrates that m6A is a potential intervention target for alleviating the adverse effects of nTiO2-induced acute pulmonary injury in vitro and in vivo, which has far-reaching implications for protecting human health and improving the sustainability of nanotechnology.
Collapse
Affiliation(s)
- Fengkai Ruan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China
| | - Changqian Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yi Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China
| | - Xisen Cao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China
| | - Zhen Tang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China
| | - Jiaying Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China
| | - Jie Zeng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China
| | - Hanying Yin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China
| | - Naying Zheng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chunyan Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361005, China.
| |
Collapse
|
9
|
Anti-Müllerian Hormone and Polycystic Ovary Syndrome in Women and Its Male Equivalent. Biomedicines 2022; 10:biomedicines10102506. [PMID: 36289767 PMCID: PMC9599141 DOI: 10.3390/biomedicines10102506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
This article reviews the main findings on anti-Müllerian hormone (AMH) and its involvement in the pathogenesis of polycystic ovary syndrome (PCOS) and its male equivalent. In women, AMH is produced by granulosa cells from the mid-fetal life to menopause and is a reliable indirect marker of ovarian reserve. AMH protects follicles from atresia, inhibits their differentiation in the ovary, and stimulates gonadotrophin-releasing hormone neurons pulsatility. AMH overexpression in women with PCOS likely contributes to the increase of the follicle cohort and of androgen levels, leading to follicular arrest and anovulation. In the male, AMH is synthesized at high levels by Sertoli cells from fetal life to puberty when serum AMH falls to levels similar to those observed in women. AMH is involved in the differentiation of the genital tract during fetal life and plays a role in Sertoli and Leydig cells differentiation and function. Serum AMH is used to assess Sertoli cell function in children with disorders of sex development and various conditions affecting the hypothalamic–pituitary–testicular axis. Although the reproductive function of male relative of women with PCOS has been poorly investigated, adolescents have elevated levels of AMH which could play a detrimental role on their fertility.
Collapse
|
10
|
Mohamed AA, Al-Hussaini TK, Hussein RS, Abdallah KS, Amer SA. The Impact of High Circulating Anti-Müllerian Hormone on Endometrial Thickness and Outcome of Assisted Reproductive Technology in Women with Polycystic Ovarian Syndrome: A Cohort Study. J Hum Reprod Sci 2022; 15:370-376. [PMID: 37033142 PMCID: PMC10077748 DOI: 10.4103/jhrs.jhrs_112_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/27/2022] [Accepted: 11/27/2022] [Indexed: 04/11/2023] Open
Abstract
Background Elevated circulating anti-Müllerian hormone (AMH) in women with the polycystic ovarian syndrome (PCOS) has been found to have a detrimental effect on endometrial function. This may adversely affect the outcome of in vitro fertilisation (IVF) in PCOS women. Aims To investigate the impact of high serum AMH concentrations on endometrial thickness (ET) and the outcome of IVF in women with PCOS. Settings and Design This retrospective cohort study included all PCOS women who underwent fresh IVF\intracytoplasmic sperm injection cycles between January 2016 and December 2021 in one major IVF centre. Materials and Methods PCOS diagnosis was based on Rotterdam criteria, and participants were identified from centre database. All women received antagonist protocol. Primary outcomes were trigger-day ET and live birth rate (LBR). Circulating AMH was correlated with ET and ovarian response. Statistical Analysis Used AMH levels were compared between women with and without live birth. ET and LBRs were compared between women with AMH <7.0 ng/ml versus those with AMH ≥7.0 ng/ml. Results The study included 102 PCOS women, of which six were excluded due to poor response (n = 4), hyperresponse (n = 1) or fertilisation failure (n = 1). Of the remaining 96 women, 42 (43.8%) achieved a live birth. There was no statistically significant (P > 0.05) correlation between AMH and ET. Mean ± standard deviation AMH concentration was not significantly (P > 0.05) different between women with live birth (6.5 ± 3.4 ng/ml) and those without (6.5 ± 2.4 ng/ml). High AMH positively correlated with the number of oocytes retrieved, metaphase II oocytes and embryos (P = 0.003, 0.006 and 0.006, respectively). There was no statistically significant (P > 0.05) difference in ET or LBR between women with AMH <7.0 ng/ml (n = 72; ET, 10.7 ± 1.8 mm; LBR, 45.8% [33/72] versus those with AMH ≥7.0 ng/ml (n = 24; ET, 10.8 ± 1.7 mm; LBR, 37.5% [9/24]). Conclusions High circulating AMH in PCOS women does not seem to negatively affect ET or LBRs during assisted reproductive technology.
Collapse
Affiliation(s)
| | | | - Reda S. Hussein
- Department of Obstetrics and Gynaecology, Assiut University, Assiut, Egypt
| | - Karim S. Abdallah
- Department of Obstetrics and Gynaecology, Assiut University, Assiut, Egypt
| | - Saad A. Amer
- Department of Obstetrics and Gynaecology, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, UK
| |
Collapse
|
11
|
Jiang W, Xie N, Xu C. Characterization of a prognostic model for lung squamous cell carcinoma based on eight stemness index-related genes. BMC Pulm Med 2022; 22:224. [PMID: 35676660 PMCID: PMC9178800 DOI: 10.1186/s12890-022-02011-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/11/2022] [Indexed: 11/17/2022] Open
Abstract
Background Cancer stem cells (CSCs) are implicated in cancer progression, chemoresistance, and poor prognosis; thus, they may be promising therapeutic targets. In this study, we aimed to investigate the prognostic application of differentially expressed CSC-related genes in lung squamous cell carcinoma (LUSC). Methods The mRNA stemness index (mRNAsi)-related differentially expressed genes (DEGs) in tumors were identified and further categorized by LASSO Cox regression analysis and 1,000-fold cross-validation, followed by the construction of a prognostic score model for risk stratification. The fractions of tumor-infiltrating immune cells and immune checkpoint genes were analyzed in different risk groups. Results We found 404 mRNAsi-related DEGs in LUSC, 77 of which were significantly associated with overall survival. An eight-gene prognostic signature (PPP1R27, TLX2, ANKLE1, TIGD3, AMH, KCNK3, FLRT3, and PPBP) was identified and used to construct a risk score model. The TCGA set was dichotomized into two risk groups that differed significantly (p = 0.00057) in terms of overall survival time (1, 3, 5-year AUC = 0.830, 0.749, and 0.749, respectively). The model performed well in two independent GEO datasets (p = 0.029, 0.033; 1-year AUC = 0747, 0.783; 3-year AUC = 0.746, 0.737; 5-year AUC = 0.706, 0.723). Low-risk patients had markedly increased numbers of CD8+ T cells and M1 macrophages and downregulated immune checkpoint genes compared to the corresponding values in high-risk patients (p < 0.05). Conclusion A stemness-related prognostic model based on eight prognostic genes in LUSC was developed and validated. The results of this study would have prognostic and therapeutic implications. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-02011-0.
Collapse
Affiliation(s)
- Wenfa Jiang
- Thoracic Surgery Department, Ganzhou People's Hospital, 16 MeiGuan Ave, Zhanggong, 341000, Ganzhou, China
| | - Ning Xie
- Thoracic Surgery Department, Ganzhou People's Hospital, 16 MeiGuan Ave, Zhanggong, 341000, Ganzhou, China
| | - Chenyang Xu
- Thoracic Surgery Department, Ganzhou People's Hospital, 16 MeiGuan Ave, Zhanggong, 341000, Ganzhou, China.
| |
Collapse
|
12
|
Joshi P, Basso B, Wang H, Hong SH, Giardina C, Shin DG. rPAC: Route based pathway analysis for cohorts of gene expression data sets. Methods 2022; 198:76-87. [PMID: 34628030 PMCID: PMC8792230 DOI: 10.1016/j.ymeth.2021.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/09/2021] [Accepted: 10/04/2021] [Indexed: 02/03/2023] Open
Abstract
Pathway analysis is a popular method aiming to derive biological interpretation from high-throughput gene expression studies. However, existing methods focus mostly on identifying which pathway or pathways could have been perturbed, given differential gene expression patterns. In this paper, we present a novel pathway analysis framework, namely rPAC, which decomposes each signaling pathway route into two parts, the upstream portion of a transcription factor (TF) block and the downstream portion from the TF block and generates a pathway route perturbation analysis scheme examining disturbance scores assigned to both parts together. This rPAC scoring is further applied to a cohort of gene expression data sets which produces two summary metrics, "Proportion of Significance" (PS) and "Average Route Score" (ARS), as quantitative measures discerning perturbed pathway routes within and/or between cohorts. To demonstrate rPAC's scoring competency, we first used a large amount of simulated data and compared the method's performance against those by conventional methods in terms of power curve. Next, we performed a case study involving three epithelial cancer data sets from The Cancer Genome Atlas (TCGA). The rPAC method revealed specific pathway routes as potential cancer type signatures. A deeper pathway analysis of sub-groups (i.e., age groups in COAD or cancer sub-types in BRCA) resulted in pathway routes that are known to be associated with the sub-groups. In addition, multiple previously uncharacterized pathways routes were identified, potentially suggesting that rPAC is better in deciphering etiology of a disease than conventional methods particularly in isolating routes and sections of perturbed pathways in a finer granularity.
Collapse
Affiliation(s)
- Pujan Joshi
- Computer Science and Engineering Department, University of Connecticut, Storrs, CT, USA.
| | - Brent Basso
- Molecular and Cell Biology Department, University of Connecticut, Storrs, CT, USA
| | - Honglin Wang
- Computer Science and Engineering Department, University of Connecticut, Storrs, CT, USA
| | - Seung-Hyun Hong
- Computer Science and Engineering Department, University of Connecticut, Storrs, CT, USA
| | - Charles Giardina
- Molecular and Cell Biology Department, University of Connecticut, Storrs, CT, USA
| | - Dong-Guk Shin
- Computer Science and Engineering Department, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
13
|
Cate RL. Anti-Müllerian Hormone Signal Transduction involved in Müllerian Duct Regression. Front Endocrinol (Lausanne) 2022; 13:905324. [PMID: 35721723 PMCID: PMC9201060 DOI: 10.3389/fendo.2022.905324] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Over seventy years ago it was proposed that the fetal testis produces a hormone distinct from testosterone that is required for complete male sexual development. At the time the hormone had not yet been identified but was invoked by Alfred Jost to explain why the Müllerian duct, which develops into the female reproductive tract, regresses in the male fetus. That hormone, anti-Müllerian hormone (AMH), and its specific receptor, AMHR2, have now been extensively characterized and belong to the transforming growth factor-β families of protein ligands and receptors involved in growth and differentiation. Much is now known about the downstream events set in motion after AMH engages AMHR2 at the surface of specific Müllerian duct cells and initiates a cascade of molecular interactions that ultimately terminate in the nucleus as activated transcription factors. The signals generated by the AMH signaling pathway are then integrated with signals coming from other pathways and culminate in a complex gene regulatory program that redirects cellular functions and fates and leads to Müllerian duct regression.
Collapse
|
14
|
Bertho S, Neyroud AS, Brun T, Jaillard S, Bonnet F, Ravel C. Anti-Müllerian hormone: A function beyond the Müllerian structures. Morphologie 2021; 106:252-259. [PMID: 34924282 DOI: 10.1016/j.morpho.2021.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 10/19/2022]
Abstract
The anti-Müllerian hormone (AMH) is a heterodimeric glycoprotein belonging to the TGFb superfamily implicated in human embryonic development. This hormone was first described as allowing regression of the epithelial embryonic Müllerian structures in males, which would otherwise differentiate into the uterus and fallopian tubes. It activates a signaling pathway mediated by two transmembrane receptors. Binding of AMH to its receptor induces morphological changes leading to the degeneration of Müllerian ducts. Recently, new data has shown the role played by this hormone on structures other than the genital tract. If testicular AMH expression decreases in humans over the course of a lifetime, synthesis may persist in other tissues in adulthood. The mechanisms underlying its production have been unveiled. The aim of this review is to describe the different pathways in which AMH has been identified and plays a pivotal role.
Collapse
Affiliation(s)
- S Bertho
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France.
| | - A S Neyroud
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France; Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France
| | - T Brun
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France
| | - S Jaillard
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France; Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France
| | - F Bonnet
- CHU Rennes, Service d'Endocrinologie, 35000 Rennes, France
| | - C Ravel
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France; Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France
| |
Collapse
|
15
|
ERK Phosphorylation Regulates the Aml1/Runx1 Splice Variants and the TRP Channels Expression during the Differentiation of Glioma Stem Cell Lines. Cells 2021; 10:cells10082052. [PMID: 34440820 PMCID: PMC8391729 DOI: 10.3390/cells10082052] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/15/2022] Open
Abstract
The identification of cancer stem cells in brain tumors paved the way for new therapeutic approaches. Recently, a role for the transcriptional factor Runx1/Aml1 and the downstream ion channel genes in brain cancer development and progression has been suggested. This study aimed to explore the expression and the role of Runx1/Aml1, its Aml1b and Aml1c splice variants and the downstream TRPA1 and TRPV1 ion channels in undifferentiated and day-14 differentiated neural stem cells (NSCs and D-NSCs) and glioblastoma stem cells (GSCs and D-GSCs) lines with different proneural (PN) or mesenchymal (MES) phenotype. Gene and protein expression were evaluated by qRT-PCR, cytofluorimetric, western blot and confocal microscopy analyses. Moreover, by western blot, we observed that ERK phosphorylation enhances the Aml1b and Aml1c protein expression during glioma differentiation. Furthermore, the agonists of TRPA1 and TRPV1 channels stimulated apoptosis/necrosis in GSCs and D-GSCs as evaluated by Annexin V and PI staining and cytofluorimetric analysis. Finally, by qRT-PCR, the modulation of Wnt/β catenin, FGF, and TGFβ/SMAD signaling pathways in PN- and MES-GSCs was reported. Overall, our results provide new evidence regarding Runx1/Aml1 isoform overexpression and modulation in TRP channel expression during gliomagenesis, thus offering new directions for glioblastoma therapy.
Collapse
|
16
|
Chauvin M, Garambois V, Choblet S, Colombo PE, Chentouf M, Gros L, De Brauwere DP, Duonor-Cerutti M, Dumas K, Robert B, Jarlier M, Martineau P, Navarro-Teulon I, Pépin D, Chardès T, Pèlegrin A. Anti-Müllerian hormone concentration regulates activin receptor-like kinase-2/3 expression levels with opposing effects on ovarian cancer cell survival. Int J Oncol 2021; 59:43. [PMID: 34013359 PMCID: PMC8131086 DOI: 10.3892/ijo.2021.5223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/11/2021] [Indexed: 11/27/2022] Open
Abstract
Anti‑Müllerian hormone (AMH) type II receptor (AMHRII) and the AMH/AMHRII signaling pathway are potential therapeutic targets in ovarian carcinoma. Conversely, the role of the three AMH type I receptors (AMHRIs), namely activin receptor‑like kinase (ALK)2, ALK3 and ALK6, in ovarian cancer remains to be clarified. To determine the respective roles of these three AMHRIs, the present study used four ovarian cancer cell lines (COV434‑AMHRII, SKOV3‑AMHRII, OVCAR8, KGN) and primary cells isolated from tumor ascites from patients with ovarian cancer. The results demonstrated that ALK2 and ALK3 may be the two main AMHRIs involved in AMH signaling at physiological endogenous and supraphysiological exogenous AMH concentrations, respectively. Supraphysiological AMH concentrations (25 nM recombinant AMH) were associated with apoptosis in all four cell lines and decreased clonogenic survival in COV434‑AMHRII and SKOV3‑AMHRII cells. These biological effects were induced via ALK3 recruitment by AMHRII, as ALK3‑AMHRII dimerization was favored at increasing AMH concentrations. By contrast, ALK2 was associated with AMHRII at physiological endogenous concentrations of AMH (10 pM). Based on these results, tetravalent IgG1‑like bispecific antibodies (BsAbs) against AMHRII and ALK2, and against AMHRII and ALK3 were designed and evaluated. In vivo, COV434‑AMHRII tumor cell xenograft growth was significantly reduced in all BsAb‑treated groups compared with that in the vehicle group (P=0.018 for BsAb 12G4‑3D7; P=0.001 for all other BsAbs). However, the growth of COV434‑AMHRII tumor cell xenografts was slower in mice treated with the anti‑AMRII‑ALK2 BsAb 12G4‑2F9 compared with that in animals that received a control BsAb that targeted AMHRII and CD5 (P=0.048). These results provide new insights into type I receptor specificity in AMH signaling pathways and may lead to an innovative therapeutic approach to modulate AMH signaling using anti‑AMHRII/anti‑AMHRI BsAbs.
Collapse
Affiliation(s)
- Maëva Chauvin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
| | - Véronique Garambois
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
| | - Sylvie Choblet
- CNRS UPS3044 Baculovirus et Thérapie, F-30380 Saint-Christol-Lèz Alès, France
| | - Pierre-Emmanuel Colombo
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, F-34298 Montpellier, France
| | - Myriam Chentouf
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
| | - Laurent Gros
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
| | | | | | | | - Bruno Robert
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
| | - Marta Jarlier
- Institut Régional du Cancer de Montpellier, ICM, F-34298 Montpellier, France
| | - Pierre Martineau
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
| | - Isabelle Navarro-Teulon
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
| | - David Pépin
- Department of Surgery, Harvard Medical School, Boston, MA 02114, USA
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Thierry Chardès
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
| | - André Pèlegrin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
| |
Collapse
|
17
|
Prat M, Salon M, Allain T, Dubreuil O, Noël G, Preisser L, Jean B, Cassard L, Lemée F, Tabah-Fish I, Pipy B, Jeannin P, Prost JF, Barret JM, Coste A. Murlentamab, a Low Fucosylated Anti-Müllerian Hormone Type II Receptor (AMHRII) Antibody, Exhibits Anti-Tumor Activity through Tumor-Associated Macrophage Reprogrammation and T Cell Activation. Cancers (Basel) 2021; 13:cancers13081845. [PMID: 33924378 PMCID: PMC8070390 DOI: 10.3390/cancers13081845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/19/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary AMHRII, the anti-Müllerian hormone receptor, is selectively expressed in normal sexual organs in healthy adults but is also re-expressed in ovarian, colorectal and lung cancers. In this context, we developed murlentamab, a humanized glyco-engineered anti-AMHRII monoclonal antibody, currently in clinical trial. Preliminary data suggest that murlentamab anti-tumor activity involves immune response activation. Thus, in vitro experiments were performed to precisely characterize the murlentamab effect on the human immune system. We show that murlentamab treatment is associated with evidences of innate and adaptive immune cell activation in cancer patient samples. Moreover, we demonstrate that the murlentamab opsonization of AMHRII-expressing ovarian tumor cells promotes a polarization switch of both naïve and tumor-associated macrophages towards an anti-tumor M1-like phenotype. Our work also supports that, through macrophage reeducation, murlentamab activates an anti-tumor adaptive immune response. Finally, the combination of murlentamab with pembrolizumab confirmed novel clinical perspectives of murlentamab association with checkpoint inhibitors and other immuno-modulators. Abstract AMHRII, the anti-Müllerian hormone receptor, is selectively expressed in normal sexual organs but is also re-expressed in gynecologic cancers. Hence, we developed murlentamab, a humanized glyco-engineered anti-AMHRII monoclonal antibody currently in clinical trial. Low-fucosylated antibodies are known to increase the antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) potency of effector cells, but some preliminary results suggest a more global murlentamab-dependent activation of the immune system. In this context, we demonstrate here that the murlentamab opsonization of AMHRII-expressing ovarian tumor cells, in the presence of unstimulated- or tumor-associated macrophage (TAM)-like macrophages, significantly promotes macrophage-mediated ADCC and shifts the whole microenvironment towards a pro-inflammatory and anti-tumoral status, thus triggering anti-tumor activity. We also report that murlentamab orients both unstimulated- and TAM-like macrophages to an M1-like phenotype characterized by a strong expression of co-stimulation markers, pro-inflammatory cytokines and chemokines, favoring T cell recruitment and activation. Moreover, we show that murlentamab treatment shifts CD4+ Th1/Th2 balance towards a Th1 response and activates CD8+ T cells. Altogether, these results suggest that murlentamab, through naïve macrophage orientation and TAM reprogrammation, stimulates the anti-tumor adaptive immune response. Those mechanisms might contribute to the sustained clinical benefit observed in advanced cancer patients treated with murlentamab. Finally, the enhanced murlentamab activity in combination with pembrolizumab opens new therapeutic perspectives.
Collapse
Affiliation(s)
- Mélissa Prat
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
| | - Marie Salon
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
| | - Thibault Allain
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
| | - Olivier Dubreuil
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Grégory Noël
- Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium;
| | - Laurence Preisser
- Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, 49000 Angers, France; (L.P.); (P.J.)
| | - Bérangère Jean
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Lydie Cassard
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94905 Villejuif, France;
| | - Fanny Lemée
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Isabelle Tabah-Fish
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Bernard Pipy
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
| | - Pascale Jeannin
- Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, 49000 Angers, France; (L.P.); (P.J.)
| | - Jean-François Prost
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Jean-Marc Barret
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Agnès Coste
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
- Correspondence: ; Tel.: +33-534-609-501
| |
Collapse
|
18
|
The Expression of Anti-Müllerian Hormone Type II Receptor (AMHRII) in Non-Gynecological Solid Tumors Offers Potential for Broad Therapeutic Intervention in Cancer. BIOLOGY 2021; 10:biology10040305. [PMID: 33917111 PMCID: PMC8067808 DOI: 10.3390/biology10040305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 11/17/2022]
Abstract
Simple Summary Until now, only a few studies have examined the AMHRII expression in tumors. Here, with more than 1000 tumor samples and using several complementary techniques we confirmed AMHRII expression in gynecological cancer and demonstrated AMHRII expression in certain non-gynecological cancers such as colorectal cancers. These findings open the way for new therapeutic approaches targeting AMHRII and emphasize the need to better understand the role of AMH/AMHRII in cancer. Abstract The anti-Müllerian hormone (AMH) belongs to the TGF-β family and plays a key role during fetal sexual development. Various reports have described the expression of AMH type II receptor (AMHRII) in human gynecological cancers including ovarian tumors. According to qRT-PCR results confirmed by specific In-Situ Hybridization (ISH) experiments, AMHRII mRNA is expressed in an extremely restricted number of normal tissues. By performing ISH on tissue microarray of solid tumor samples AMHRII mRNA was unexpectedly detected in several non-gynecological primary cancers including lung, breast, head and neck, and colorectal cancers. AMHRII protein expression, evaluated by immunohistochemistry (IHC) was detected in approximately 70% of epithelial ovarian cancers. Using the same IHC protocol on more than 900 frozen samples covering 18 different cancer types we detected AMHRII expression in more than 50% of hepato-carcinomas, colorectal, lung, and renal cancer samples. AMHRII expression was not observed in neuroendocrine lung tumor samples nor in non-Hodgkin lymphoma samples. Complementary analyses by immunofluorescence and flow cytometry confirmed the detection of AMHRII on a panel of ovarian and colorectal cancers displaying comparable expression levels with mean values of 39,000 and 50,000 AMHRII receptors per cell, respectively. Overall, our results suggest that this embryonic receptor could be a suitable target for treating AMHRII-expressing tumors with an anti-AMHRII selective agent such as murlentamab, also named 3C23K or GM102. This potential therapeutic intervention was confirmed in vivo by showing antitumor activity of murlentamab against AMHRII-expressing colorectal cancer and hepatocarcinoma Patient-Derived tumor Xenografts (PDX) models.
Collapse
|
19
|
Rak AY, Trofimov AV, Ischenko AM, Sokolov AV. [The study of interaction of different forms of human recombinant anti-mullerian hormone with a chimeric analogue of the AMH type II]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 67:66-73. [PMID: 33645523 DOI: 10.18097/pbmc20216701066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The homodimeric glycoprotein, anti-mullerian hormone (AMH), described over 70 years ago by A. Jost, is the least studied member of the transforming growth factor beta superfamily. Despite the antitumor activity of AMH discovered at the end of the last century, the creation of effective drugs based on AMH is hindered primarily by the lack of information on the mechanism of various AMH forms interaction with a specific type II receptor (MISRII). Previously, we have shown that not only the full-length activated hormone but also its C-terminal fragment (C-rAMH) could bind to MISRII. In this work, using the surface plasmon resonance technique, we compared the interaction of three forms of recombinant AMH (rAMH) with the MISRII analogue - the chimeric protein MISRII-Fc containing AMH type II receptor and a Fc-fragment of the human IgG1 heavy chain. Comparison of the binding of MISRII-Fc, immobilized on a chip with group specificity for human immunoglobulins, to C-rAMH, to intact rAMH (pro-rAMH), and to rAMH containing one uncleaved monomer (hc-rAMH), showed that the KD of the complexes increased: 1.7 nM, 88 nM and 110 nM, respectively. Thus, we have shown that C-terminal fragment of AMH has the maximum affinity for the recombinant MISRII analogue, which indicates the prospects for the development of drugs based on this hormone derivative.
Collapse
Affiliation(s)
- A Ya Rak
- State Research Institute for Highly Pure Biopreparations, Saint-Petersburg, Russia
| | - A V Trofimov
- State Research Institute for Highly Pure Biopreparations, Saint-Petersburg, Russia
| | - A M Ischenko
- State Research Institute for Highly Pure Biopreparations, Saint-Petersburg, Russia
| | - A V Sokolov
- Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
20
|
Chauvin M, Garambois V, Colombo PE, Chentouf M, Gros L, Brouillet JP, Robert B, Jarlier M, Dumas K, Martineau P, Navarro-Teulon I, Pépin D, Chardès T, Pèlegrin A. Anti-Müllerian hormone (AMH) autocrine signaling promotes survival and proliferation of ovarian cancer cells. Sci Rep 2021; 11:2231. [PMID: 33500516 PMCID: PMC7838181 DOI: 10.1038/s41598-021-81819-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/12/2021] [Indexed: 12/30/2022] Open
Abstract
In ovarian carcinoma, anti-Müllerian hormone (AMH) type II receptor (AMHRII) and the AMH/AMHRII signaling pathway are potential therapeutic targets. Here, AMH dose-dependent effect on signaling and proliferation was analyzed in four ovarian cancer cell lines, including sex cord stromal/granulosa cell tumors and high grade serous adenocarcinomas (COV434-AMHRII, SKOV3-AMHRII, OVCAR8 and KGN). As previously shown, incubation with exogenous AMH at concentrations above the physiological range (12.5-25 nM) decreased cell viability. Conversely, physiological concentrations of endogenous AMH improved cancer cell viability. Partial AMH depletion by siRNAs was sufficient to reduce cell viability in all four cell lines, by 20% (OVCAR8 cells) to 40% (COV434-AMHRII cells). In the presence of AMH concentrations within the physiological range (5 to 15 pM), the newly developed anti-AMH B10 antibody decreased by 25% (OVCAR8) to 50% (KGN) cell viability at concentrations ranging between 3 and 333 nM. At 70 nM, B10 reduced clonogenic survival by 57.5%, 57.1%, 64.7% and 37.5% in COV434-AMHRII, SKOV3-AMHRII, OVCAR8 and KGN cells, respectively. In the four cell lines, B10 reduced AKT phosphorylation, and increased PARP and caspase 3 cleavage. These results were confirmed in ovarian cancer cells isolated from patients' ascites, demonstrating the translational potential of these results. Furthermore, B10 reduced COV434-MISRII tumor growth in vivo and significantly enhanced the median survival time compared with vehicle (69 vs 60 days; p = 0.0173). Our data provide evidence for a novel pro-survival autocrine role of AMH in the context of ovarian cancer, which was targeted therapeutically using an anti-AMH antibody to successfully repress tumor growth.
Collapse
Affiliation(s)
- Maëva Chauvin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France
- INSERM, U1194, 34298, Montpellier, France
- Université de Montpellier, 34298, Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - Véronique Garambois
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France
- INSERM, U1194, 34298, Montpellier, France
- Université de Montpellier, 34298, Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France
| | - Pierre-Emmanuel Colombo
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France
- INSERM, U1194, 34298, Montpellier, France
- Université de Montpellier, 34298, Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France
| | - Myriam Chentouf
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France
- INSERM, U1194, 34298, Montpellier, France
- Université de Montpellier, 34298, Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France
| | - Laurent Gros
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France
- INSERM, U1194, 34298, Montpellier, France
- Université de Montpellier, 34298, Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France
| | - Jean-Paul Brouillet
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France
- INSERM, U1194, 34298, Montpellier, France
- Université de Montpellier, 34298, Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France
- Département de Biochimie et Biologie Moléculaire, CHU de Nîmes, Nîmes, France
| | - Bruno Robert
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France
- INSERM, U1194, 34298, Montpellier, France
- Université de Montpellier, 34298, Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France
| | - Marta Jarlier
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France
- INSERM, U1194, 34298, Montpellier, France
- Université de Montpellier, 34298, Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France
| | - Karen Dumas
- SurgiMAb, 10 Parc Club du Millénaire, 1025 Avenue Henri Becquerel, 34000, Montpellier, France
| | - Pierre Martineau
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France
- INSERM, U1194, 34298, Montpellier, France
- Université de Montpellier, 34298, Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France
| | - Isabelle Navarro-Teulon
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France
- INSERM, U1194, 34298, Montpellier, France
- Université de Montpellier, 34298, Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France
| | - David Pépin
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - Thierry Chardès
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France
- INSERM, U1194, 34298, Montpellier, France
- Université de Montpellier, 34298, Montpellier, France
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France
| | - André Pèlegrin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Campus Val d'Aurelle, 34298, Montpellier Cedex, France.
- INSERM, U1194, 34298, Montpellier, France.
- Université de Montpellier, 34298, Montpellier, France.
- Institut Régional du Cancer de Montpellier, ICM, 34298, Montpellier, France.
| |
Collapse
|
21
|
Palomba S, Piltonen TT, Giudice LC. Endometrial function in women with polycystic ovary syndrome: a comprehensive review. Hum Reprod Update 2020; 27:584-618. [PMID: 33302299 DOI: 10.1093/humupd/dmaa051] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/29/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility. An endometrial component has been suggested to contribute to subfertility and poor reproductive outcomes in affected women. OBJECTIVE AND RATIONALE The aim of this review was to determine whether there is sufficient evidence to support that endometrial function is altered in women with PCOS, whether clinical features of PCOS affect the endometrium, and whether there are evidence-based interventions to improve endometrial dysfunction in PCOS women. SEARCH METHODS An extensive literature search was performed from 1970 up to July 2020 using PubMed and Web of Science without language restriction. The search included all titles and abstracts assessing a relationship between PCOS and endometrial function, the role played by clinical and biochemical/hormonal factors related to PCOS and endometrial function, and the potential interventions aimed to improve endometrial function in women with PCOS. All published papers were included if considered relevant. Studies having a specific topic/hypothesis regarding endometrial cancer/hyperplasia in women with PCOS were excluded from the analysis. OUTCOMES Experimental and clinical data suggest that the endometrium differs in women with PCOS when compared to healthy controls. Clinical characteristics related to the syndrome, alone and/or in combination, may contribute to dysregulation of endometrial expression of sex hormone receptors and co-receptors, increase endometrial insulin-resistance with impaired glucose transport and utilization, and result in chronic low-grade inflammation, immune dysfunction, altered uterine vascularity, abnormal endometrial gene expression and cellular abnormalities in women with PCOS. Among several interventions to improve endometrial function in women with PCOS, to date, only lifestyle modification, metformin and bariatric surgery have the highest scientific evidence for clinical benefit. WIDER IMPLICATIONS Endometrial dysfunction and abnormal trophoblast invasion and placentation in PCOS women can predispose to miscarriage and pregnancy complications. Thus, patients and their health care providers should advise about these risks. Although currently no intervention can be universally recommended to reverse endometrial dysfunction in PCOS women, lifestyle modifications and metformin may improve underlying endometrial dysfunction and pregnancy outcomes in obese and/or insulin resistant patients. Bariatric surgery has shown its efficacy in severely obese PCOS patients, but a careful evaluation of the benefit/risk ratio is warranted. Large scale randomized controlled clinical trials should address these possibilities.
Collapse
Affiliation(s)
- Stefano Palomba
- Unit of Obstetrics and Gynecology, Grande Ospedale Metropolitano of Reggio Calabria, Reggio Calabria, Italy
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Linda C Giudice
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| |
Collapse
|
22
|
Fogg KC, Miller AE, Li Y, Flanigan W, Walker A, O'Shea A, Kendziorski C, Kreeger PK. Ovarian cancer cells direct monocyte differentiation through a non-canonical pathway. BMC Cancer 2020; 20:1008. [PMID: 33069212 PMCID: PMC7568422 DOI: 10.1186/s12885-020-07513-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 10/08/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Alternatively-activated macrophages (AAMs), an anti-inflammatory macrophage subpopulation, have been implicated in the progression of high grade serous ovarian carcinoma (HGSOC). Increased levels of AAMs are correlated with poor HGSOC survival rates, and AAMs increase the attachment and spread of HGSOC cells in vitro. However, the mechanism by which monocytes in the HGSOC tumor microenvironment are differentiated and polarized to AAMs remains unknown. METHODS Using an in vitro co-culture device, we cultured naïve, primary human monocytes with a panel of five HGSOC cell lines over the course of 7 days. An empirical Bayesian statistical method, EBSeq, was used to couple RNA-seq with observed monocyte-derived cell phenotype to explore which HGSOC-derived soluble factors supported differentiation to CD68+ macrophages and subsequent polarization towards CD163+ AAMs. Pathways of interest were interrogated using small molecule inhibitors, neutralizing antibodies, and CRISPR knockout cell lines. RESULTS HGSOC cell lines displayed a wide range of abilities to generate AAMs from naïve monocytes. Much of this variation appeared to result from differential ability to generate CD68+ macrophages, as most CD68+ cells were also CD163+. Differences in tumor cell potential to generate macrophages was not due to a MCSF-dependent mechanism, nor variance in established pro-AAM factors. TGFα was implicated as a potential signaling molecule produced by tumor cells that could induce macrophage differentiation, which was validated using a CRISPR knockout of TGFA in the OVCAR5 cell line. CONCLUSIONS HGSOC production of TGFα drives monocytes to differentiate into macrophages, representing a central arm of the mechanism by which AAMs are generated in the tumor microenvironment.
Collapse
Affiliation(s)
- Kaitlin C Fogg
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave, WIMR 4553, Madison, WI, 53705, USA
| | - Andrew E Miller
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave, WIMR 4553, Madison, WI, 53705, USA
| | - Ying Li
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Will Flanigan
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave, WIMR 4553, Madison, WI, 53705, USA
| | - Alyssa Walker
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave, WIMR 4553, Madison, WI, 53705, USA
| | - Andrea O'Shea
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Pamela K Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave, WIMR 4553, Madison, WI, 53705, USA.
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
23
|
Nasrin Ferdousy R, Kereilwe O, Kadokawa H. Anti-Müllerian hormone is expressed and secreted by bovine oviductal and endometrial epithelial cells. Anim Sci J 2020; 91:e13456. [PMID: 32926548 DOI: 10.1111/asj.13456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 11/29/2022]
Abstract
In this study, we investigated whether bovine oviducts and endometria produce anti-Müllerian hormone (AMH) (for paracrine and autocrine signaling). Reverse transcription-polymerase chain reaction and western blotting detected AMH expression in oviductal and endometrial specimens. Immunohistochemistry revealed robust AMH expression in the ampulla and isthmus epithelia, and the glandular and luminal endometrial epithelia (caruncular endometria). AMH mRNA (measured by real-time PCR) and protein expression in these layers did not significantly differ among estrous phases in adult Japanese Black (JB) heifers (p > .1). Furthermore, the expression in these layers also did not differ among Holstein cows (93.8 ± 5.8 months old), JB heifers (25.5 ± 0.4 months old), and JB cows (97.9 ± 7.9 months old). We also compared AMH concentrations in the oviduct and uterine horn fluids among the three groups (measured by immunoassays). Interestingly, the AMH concentration in the oviduct fluid, but not in the uterine horn fluid, of Holstein cows was lower than those in JB heifers and cows (p < .05). Therefore, bovine oviducts and endometria express AMH and likely secrete it into the oviduct and uterine fluids.
Collapse
Affiliation(s)
| | - Onalenna Kereilwe
- Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Hiroya Kadokawa
- Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
24
|
Li R, Liu X, Zhou XJ, Chen X, Li JP, Yin YH, Qu YQ. Identification of a Prognostic Model Based on Immune-Related Genes of Lung Squamous Cell Carcinoma. Front Oncol 2020; 10:1588. [PMID: 33014809 PMCID: PMC7493716 DOI: 10.3389/fonc.2020.01588] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/23/2020] [Indexed: 12/23/2022] Open
Abstract
Immune-related genes (IRGs) play considerable roles in tumor immune microenvironment (IME). This research aimed to discover the differentially expressed immune-related genes (DEIRGs) based on the Cox predictive model to predict survival for lung squamous cell carcinoma (LUSC) through bioinformatics analysis. First of all, the differentially expressed genes (DEGs) were acquired based on The Cancer Genome Atlas (TCGA) using the limma R package, the DEIRGs were obtained from the ImmPort database, whereas the differentially expressed transcription factors (DETFs) were acquired from the Cistrome database. Thereafter, a TFs-mediated IRGs network was constructed to identify the candidate mechanisms for those DEIRGs in LUSC at molecular level. Moreover, Gene Ontology (GO), together with Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, was conducted for exploring those functional enrichments for DEIRGs. Besides, univariate as well as multivariate Cox regression analysis was conducted for establishing a prediction model for DEIRGs biomarkers. In addition, the relationship between the prognostic model and immunocytes was further explored through immunocyte correlation analysis. In total, 3,599 DEGs, 223 DEIRGs, and 46 DETFs were obtained from LUSC tissues and adjacent non-carcinoma tissues. According to multivariate Cox regression analysis, 10 DEIRGs (including CALCB, GCGR, HTR3A, AMH, VGF, SEMA3B, NRTN, ENG, ACVRL1, and NR4A1) were retrieved to establish a prognostic model for LUSC. Immunocyte infiltration analysis showed that dendritic cells and neutrophils were positively correlated with IRGs, which possibly exerted an important part within the IME of LUSC. Our study identifies a prognostic model based on IRGs, which is then used to predict LUSC prognosis and analyze immunocyte infiltration. This may provide a novel insight for exploring the potential IRGs in the IME of LUSC.
Collapse
Affiliation(s)
- Rui Li
- Department of Pulmonary and Critical Care Medicine, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Xiao Liu
- Department of Pulmonary and Critical Care Medicine, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Xi-Jia Zhou
- Department of Pulmonary and Critical Care Medicine, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Xiao Chen
- Department of Pulmonary and Critical Care Medicine, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China.,Department of Respiratory Medicine, Tai'an City Central Hospital, Tai'an, China
| | - Jian-Ping Li
- Department of Pulmonary and Critical Care Medicine, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Yun-Hong Yin
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yi-Qing Qu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
25
|
Gaponova AV, Rodin S, Mazina AA, Volchkov PV. Epithelial-Mesenchymal Transition: Role in Cancer Progression and the Perspectives of Antitumor Treatment. Acta Naturae 2020; 12:4-23. [PMID: 33173593 PMCID: PMC7604894 DOI: 10.32607/actanaturae.11010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
About 90% of all malignant tumors are of epithelial nature. The epithelial tissue is characterized by a close interconnection between cells through cell-cell interactions, as well as a tight connection with the basement membrane, which is responsible for cell polarity. These interactions strictly determine the location of epithelial cells within the body and are seemingly in conflict with the metastatic potential that many cancers possess (the main criteria for highly malignant tumors). Tumor dissemination into vital organs is one of the primary causes of death in patients with cancer. Tumor dissemination is based on the so-called epithelial-mesenchymal transition (EMT), a process when epithelial cells are transformed into mesenchymal cells possessing high mobility and migration potential. More and more studies elucidating the role of the EMT in metastasis and other aspects of tumor progression are published each year, thus forming a promising field of cancer research. In this review, we examine the most recent data on the intracellular and extracellular molecular mechanisms that activate EMT and the role they play in various aspects of tumor progression, such as metastasis, apoptotic resistance, and immune evasion, aspects that have usually been attributed exclusively to cancer stem cells (CSCs). In conclusion, we provide a detailed review of the approved and promising drugs for cancer therapy that target the components of the EMT signaling pathways.
Collapse
Affiliation(s)
- A. V. Gaponova
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701 Russia
| | - S. Rodin
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177 Sweden
| | - A. A. Mazina
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701 Russia
| | - P. V. Volchkov
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701 Russia
| |
Collapse
|
26
|
Mishra NK, Southekal S, Guda C. Prognostic value of biomarkers in the tumor microenvironment of pancreatic ductal adenocarcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:615. [PMID: 32566552 PMCID: PMC7290607 DOI: 10.21037/atm.2020.03.59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Nitish K Mishra
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Siddesh Southekal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
27
|
Rak AY, Trofimov AV, Pigareva NV, Protasov EA, Karabanova EA, Ischenko AM. Purification of human recombinant anti-mullerian hormone and its derivatives. Biomed Chromatogr 2020; 34:e4782. [PMID: 31845358 DOI: 10.1002/bmc.4782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/22/2019] [Accepted: 12/13/2019] [Indexed: 11/05/2022]
Abstract
Anti-mullerian hormone (AMH) is a cytokine of transforming growth factor β (TGF-β) superfamily able to induce apoptosis in cells bearing specific AMH type II receptors (AMHRII). AMHRII is overexpressed in some malignant cells, so at present recombinant AMH (rAMH) is considered as a new candidate antineoplastic drug. The use of rAMH may be especially effective in case of such severe diseases as ovarian, prostate and breast cancer. However, the development of a new drug is hampered by the laboriousness of obtaining highly purified rAMH and by the lack of data about the pharmacological characteristics of rAMH derivatives. In this work, we obtained preparations of prohormone, half-cleaved rAMH and a C-terminal fragment of rAMH, which was confirmed by qualitative and quantitative analyses. To obtain rAMH and its derivatives we used a previously developed highly effective producer strain containing the optimized human AMH gene. The production process has been divided into several stages: (a) rAMH biosynthesis in the bioreactor; (b) culture media preparation; (c) purification of rAMH and its derivatives using immunoaffinity chromatography and reversed-phase HPLC; (d) identification of the purified proteins by immunoblotting and analytical reversed-phase HPLC; and (e) evaluation of the hormone forms activity. The obtained proteins may be used in preclinical trials and in vitro study of rAMH derivatives properties.
Collapse
Affiliation(s)
- Alexandra Ya Rak
- State Research Institute of Highly Pure Biopreparations, St Petersburg, Russian Federation.,St. Petersburg State University, St Petersburg, Russian Federation
| | - Alexander V Trofimov
- State Research Institute of Highly Pure Biopreparations, St Petersburg, Russian Federation
| | - Natalia V Pigareva
- State Research Institute of Highly Pure Biopreparations, St Petersburg, Russian Federation
| | - Eugeny A Protasov
- State Research Institute of Highly Pure Biopreparations, St Petersburg, Russian Federation
| | - Elena A Karabanova
- State Research Institute of Highly Pure Biopreparations, St Petersburg, Russian Federation
| | - Alexander M Ischenko
- State Research Institute of Highly Pure Biopreparations, St Petersburg, Russian Federation
| |
Collapse
|
28
|
Exercise enhances skeletal muscle regeneration by promoting senescence in fibro-adipogenic progenitors. Nat Commun 2020; 11:889. [PMID: 32060352 PMCID: PMC7021787 DOI: 10.1038/s41467-020-14734-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 01/29/2020] [Indexed: 12/16/2022] Open
Abstract
Idiopathic inflammatory myopathies cause progressive muscle weakness and degeneration. Since high-dose glucocorticoids might not lead to full recovery of muscle function, physical exercise is also an important intervention, but some exercises exacerbate chronic inflammation and muscle fibrosis. It is unknown how physical exercise can have both beneficial and detrimental effects in chronic myopathy. Here we show that senescence of fibro-adipogenic progenitors (FAPs) in response to exercise-induced muscle damage is needed to establish a state of regenerative inflammation that induces muscle regeneration. In chronic inflammatory myopathy model mice, exercise does not promote FAP senescence or resistance against tumor necrosis factor–mediated apoptosis. Pro-senescent intervention combining exercise and pharmacological AMPK activation reverses FAP apoptosis resistance and improves muscle function and regeneration. Our results demonstrate that the absence of FAP senescence after exercise leads to muscle degeneration with FAP accumulation. FAP-targeted pro-senescent interventions with exercise and pharmacological AMPK activation may constitute a therapeutic strategy for chronic inflammatory myopathy. Some exercises exacerbate chronic inflammation and muscle fibrosis in chronic myopathy. Here, the authors show that senescence of fibro-adipogenic progenitors (FAPs) in response to exercise induces muscle regeneration, and impaired FAP senescence worsens inflammation and fibrosis in chronic myopathy in mice.
Collapse
|
29
|
The MyoD family inhibitor domain-containing protein enhances the chemoresistance of cancer stem cells in the epithelial state by increasing β-catenin activity. Oncogene 2020; 39:2377-2390. [PMID: 31911618 DOI: 10.1038/s41388-019-1152-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 12/12/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023]
Abstract
Cancer cells with mesenchymal attributes potentially display chemoresistance. Cancer stem cells (CSCs), which are intrinsically resistant to most chemotherapy agents, exhibit considerable phenotypic heterogeneity in their epithelial versus mesenchymal states. However, the drug response of CSCs in the epithelial and mesenchymal states has not been completely investigated. In this study, we found that epithelial-type (E-cadherinhigh/CD133high) CSCs displayed a higher sphere formation ability and chemoresistance than mesenchymal-type (E-cadherinlowCD133high) CSCs. Gene expression profiling of the CSC and non-CSC subpopulations with distinct epithelial-to-mesenchymal transition (EMT) states showed that MyoD family inhibitor domain-containing (MDFIC) was selectively upregulated in epithelial-type CSCs. Knockdown of MDFIC sensitized epithelial-type CSCs to chemotherapy agents. Ectopic expression of MDFIC increased the chemoresistance of mesenchymal-type CSCs. In a tissue microarray, high MDFIC expression was associated with poor prognosis of non-small cell lung cancer (NSCLC) patients. A mechanistic study showed that the MDFIC p32 isoform, which is located in the cytoplasm, interacted with the destruction complex, Axin/GSK-3/β-catenin. This interaction stabilized β-catenin by inhibiting β-catenin phosphorylation at S33/37 and increased the nuclear translocation and transcriptional activity of β-catenin. Knockdown of β-catenin decreased MDFIC-enhanced chemoresistance. These results suggested that the upregulation of MDFIC enhanced the chemoresistance of epithelial-type CSCs by elevating β-catenin activity. Thus, targeting MDFIC-regulated β-catenin signaling of epithelial-type CSCs may be a potential strategy to overcome chemoresistance in NSCLC.
Collapse
|
30
|
CAR T Cells Targeting MISIIR for the Treatment of Ovarian Cancer and Other Gynecologic Malignancies. Mol Ther 2019; 28:548-560. [PMID: 31870622 DOI: 10.1016/j.ymthe.2019.11.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 11/22/2019] [Accepted: 11/29/2019] [Indexed: 01/20/2023] Open
Abstract
The prognosis of patients diagnosed with advanced ovarian or endometrial cancer remains poor, and effective therapeutic strategies are limited. The Müllerian inhibiting substance type 2 receptor (MISIIR) is a transforming growth factor β (TGF-β) receptor family member, overexpressed by most ovarian and endometrial cancers while absent in most normal tissues. Restricted tissue expression, coupled with an understanding that MISIIR ligation transmits apoptotic signals to cancer cells, makes MISIIR an attractive target for tumor-directed therapeutics. However, the development of clinical MISIIR-targeted agents has been challenging. Prompted by the responses achieved in patients with blood malignancies using chimeric antigen receptor (CAR) T cell therapy, we hypothesized that MISIIR targeting may be achieved using a CAR T cell approach. Herein, we describe the development and evaluation of a CAR that targets MISIIR. T cells expressing the MISIIR-specific CAR demonstrated antigen-specific reactivity in vitro and eliminated MISIIR-overexpressing tumors in vivo. MISIIR CAR T cells also recognized a panel of human ovarian and endometrial cancer cell lines, and they lysed a battery of patient-derived tumor specimens in vitro, without mediating cytotoxicity of a panel of normal primary human cells. In conclusion, these results indicate that MISIIR targeting for the treatment of ovarian cancer and other gynecologic malignancies is achievable using CAR technology.
Collapse
|
31
|
Choi SH, Jung YK, Jang JA, Han S. Idiopathic pulmonary arterial hypertension associated with a novel frameshift mutation in the bone morphogenetic protein receptor II gene and enhanced bone morphogenetic protein signaling: A case report. Medicine (Baltimore) 2019; 98:e17594. [PMID: 31626133 PMCID: PMC6824762 DOI: 10.1097/md.0000000000017594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
RATIONALE Idiopathic pulmonary arterial hypertension (IPAH) is characterized by intense remodeling of small pulmonary arteries. Loss-of-function mutation of bone morphogenetic protein receptor II (BMPR2) gene and exaggerated activation of transforming growth factor (TGF)-β signaling play a critical role in this process. PATIENT CONCERNS AND DIAGNOSIS We report a novel frameshift mutation (c.117InsT, p.Y40fsX48) of the BMPR2 gene identified in a 19-year-old IPAH patient with syncope. Despite BMPR2 mutation, the phosphorylation of Smad2/3 and Samd1/5/8 was increased in the patient's peripheral blood mononuclear cells, and this event was accompanied by the upregulation of bone morphogenetic protein (BMP) signaling target genes, but not TGF-β signaling target genes. Moreover, we observed an increased expression of other BMPRs, that is, anti-Mullerian hormone type-2 receptor and the activin receptor-like kinases (ALK) 1, ALK3, and ALK6. INTERVENTIONS AND OUTCOMES The patient was prescribed a combination of macitentan, sildenafil, and nifedipine, which successfully controlled her symptom of syncope and normalized N-terminal pro-brain natriuretic peptide level after 3 months of medication. LESSONS In light of these results, we propose a new pathogenetic mechanism for IPAH, based on enhanced BMP signaling via the functional replacement of mutated BMPR2 by other BMP receptors.
Collapse
Affiliation(s)
- Sun Ha Choi
- Department of Internal medicine, Kyungpook National University Hospital, Daegu
| | - Youn-Kwan Jung
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, Gyeongsangnam-do
| | - Ji-Ae Jang
- Laboratory for arthritis and bone biology, Fatima Research Institute, Daegu Fatima Hospital, Daegu, Republic of Korea
| | - Seungwoo Han
- Department of Internal medicine, Kyungpook National University Hospital, Daegu
- Laboratory for arthritis and bone biology, Fatima Research Institute, Daegu Fatima Hospital, Daegu, Republic of Korea
| |
Collapse
|
32
|
Rak AY, Trofimov AV, Ischenko AM. Anti-mullerian hormone receptor type II as a Potential Target for Antineoplastic Therapy. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2019. [DOI: 10.1134/s1990750819030053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Rak AY, Trofimov AV, Ischenko AM. [Mullerian inhibiting substance type II receptor as a potential target for antineoplastic therapy]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 65:202-213. [PMID: 31258143 DOI: 10.18097/pbmc20196503202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The review considers properties of the type II anti-Mullerian hormone receptor (mullerian inhibiting substance receptor type II, MISRII), a transmembrane sensor with its own serine/threonine protein kinase activity, triggering apoptosis of the Mullerian ducts in mammalian embryogenesis and providing formation of the male type reproductive system. According to recent data, MISRII overexpression in the postnatal period is found in cells of a number of ovarian, mammary gland, and prostate tumors, and anti-Mullerian hormone (AMH) has a pro-apoptotic effect on MISRII-positive tumor cells. This fact makes MISRII a potential target for targeted anti-cancer therapy. Treatment based on targeting MISRII seems to be a much more effective alternative to the traditional one and will significantly reduce the drug dose. However, the mechanism of MISRII-AMH interaction is still poorly understood, so the development of new anticancer drugs is complicated. The review analyzes MISRII molecular structure and expression levels in various tissues and cell lines, as well as current understanding of the AMH binding mechanisms and data on the possibility of using MISRII as a target for the action of AMH-based antineoplastic drugs.
Collapse
Affiliation(s)
- A Ya Rak
- State Research Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia; Saint-Petersburg State University, Saint-Petersburg, Russia
| | - A V Trofimov
- State Research Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia
| | - A M Ischenko
- State Research Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia
| |
Collapse
|
34
|
Beck TN, Deneka AY, Chai L, Kanach C, Johal P, Alvarez NJ, Boumber Y, Golemis EA, Laub GW. An improved method of delivering a sclerosing agent for the treatment of malignant pleural effusion. BMC Cancer 2019; 19:614. [PMID: 31234819 PMCID: PMC6589887 DOI: 10.1186/s12885-019-5777-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/30/2019] [Indexed: 12/26/2022] Open
Abstract
Background Malignant pleural effusion (MPE) is a devastating sequela associated with cancer. Talc pleurodesis is a common treatment strategy for MPE but has been estimated to be unsuccessful in up to 20–50% of patients. Clinical failure of talc pleurodesis is thought to be due to poor dispersion. This monograph reports the development of a foam delivery system designed to more effectively coat the pleural cavity. Methods C57BL/6 mice were injected with Lewis lung carcinoma (LL/2) cells intrapleurally to induce MPE. The mice then received either normal saline (NS) control, foam control (F), talc slurry (TS, 2 mg/g) or talc foam (TF, 2 mg/g). Airspace volume was evaluated by CT, lungs/pleura were collected, and percent fibrosis was determined. Results The TF group had significantly better survival than the TS group (21 vs 13.5 days, p < 0.0001). The average effusion volume was less in the talc groups compared to the control group (140 vs 628 μL, p < 0.001). TF induced significant lung fibrosis (p < 0.01), similar to TS. On CT, TF significantly (p < 0.05) reduced loss of right lung volume (by 30–40%) compared to the control group. This was not seen with TS (p > 0.05). Conclusions This report describes using a novel talc foam delivery system for the treatment of MPE. In the LL/2 model, mice treated with the TF had better survival outcomes and less reduction of lung volume than mice treated with the standard of care TS. These data provide support for translational efforts to move talc foam from animal models into clinical trials.
Collapse
Affiliation(s)
- Tim N Beck
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA. .,Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| | - Alexander Y Deneka
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.,Department of Biochemistry, Kazan Federal University, Kazan, Russia
| | - Louis Chai
- Department of Cardiothoracic Surgery, Drexel University College of Medicine, Hahnemann University Hospital, 230 North Broad Street, Philadelphia, PA, 19102, USA
| | - Colin Kanach
- Department of Pathology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Priya Johal
- Department of Pathology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Nicolas J Alvarez
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA, 19129, USA
| | - Yanis Boumber
- Department of Biochemistry, Kazan Federal University, Kazan, Russia.,Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Glenn W Laub
- Department of Cardiothoracic Surgery, Drexel University College of Medicine, Hahnemann University Hospital, 230 North Broad Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
35
|
Beck TN, Boumber YA, Aggarwal C, Pei J, Thrash-Bingham C, Fittipaldi P, Vlasenkova R, Rao C, Borghaei H, Cristofanilli M, Mehra R, Serebriiskii I, Alpaugh RK. Circulating tumor cell and cell-free RNA capture and expression analysis identify platelet-associated genes in metastatic lung cancer. BMC Cancer 2019; 19:603. [PMID: 31215484 PMCID: PMC6582501 DOI: 10.1186/s12885-019-5795-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 06/05/2019] [Indexed: 12/25/2022] Open
Abstract
Background Circulating tumor cells (CTC) and plasma cell-free RNA (cfRNA) can serve as biomarkers for prognosis and treatment response in lung cancer. One barrier to the selected or routine use of CTCs and plasma cfRNA in precision oncology is the limited quantity of both, and CTCs are only seen in metastatic disease. As capture of CTCs and plasma cfRNA presents an opportunity to monitor and assess malignancies without invasive procedures, we compared two methods for CTC capture and identification, and profiled mRNA from CTCs and plasma cfRNA to identify potential tumor-associated biomarkers. Methods Peripheral blood was collected from ten patients with small cell lung cancer (SCLC), ten patients with non-small cell lung cancer (NSCLC) and four healthy volunteers. Two methods were used for CTC capture: the standard epithelial cell adhesion molecule (EpCam) CellSearch kit (unicapture) and EpCAM plus HER2, EGFR and MUC-1 specific combined ferrofluid capture (quadcapture). For the quadcapture, anti-cytokeratin 7 (CK7) was additionally used to assist in CTC identification. NanoString analysis was performed on plasma cfRNA and on mRNA from combined ferrofluid isolated CTCs. Expression data was analyzed using STRING and Reactome. Results Unicapture detected CTCs in 40% of NSCLC and 60% of SCLC; whereas, quadcapture/CK7 identified CTCs in 20% of NSCLC and 80% of SCLC. Bioinformatic analysis of NanoString data identified high expression of a platelet factor 4 (PF4)-related group of transcripts. Conclusions Quadcapture ferrofluid reagent did not significantly improve CTC capture efficacy. NanoString analysis based on CTC and plasma cfRNA data highlighted an intriguing PF-4-centric network in patients with metastatic lung cancer. Electronic supplementary material The online version of this article (10.1186/s12885-019-5795-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tim N Beck
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.,Digestive Disease & Surgery Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Yanis A Boumber
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.,Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.,Kazan Federal University, Kazan, Russian Federation
| | - Charu Aggarwal
- Abramson Cancer Center and Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jianming Pei
- Genomics Facility, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | | | - Patricia Fittipaldi
- Protocol Support Laboratory, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | | | - Chandra Rao
- Janssen Diagnostics LLC, Valley, Huntingdon, PA, 19006, USA
| | - Hossein Borghaei
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.,Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Massimo Cristofanilli
- Feinberg School of Medicine, Robert H Lurie Comprehensive Cancer Center, Chicago, IL, 60611, USA
| | - Ranee Mehra
- Head and Neck Medical Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| | - Ilya Serebriiskii
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.,Kazan Federal University, Kazan, Russian Federation
| | - R Katherine Alpaugh
- Protocol Support Laboratory, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA. .,Biostatistics Facility, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.
| |
Collapse
|
36
|
Beck TN, Kudinov AE, Dulaimi E, Boumber Y. Case report: reinitiating pembrolizumab treatment after small bowel perforation. BMC Cancer 2019; 19:379. [PMID: 31018834 PMCID: PMC6482547 DOI: 10.1186/s12885-019-5577-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 04/03/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have emerged as paradigm shifting treatment options for a number of cancers. Six antibodies targeting the immune checkpoint proteins programmed cell death 1 (PD-1), programmed cell death 1 ligand 1 (PD-L1) or cytotoxic T-lymphocyte associated protein 4 (CTLA4) have been approved. In some cases, response rates have been impressive, but not uniformly so and not consistently; similarly, toxicity to this class of therapeutic is often unpredictable and can be life threatening. Predicting treatment response and toxicity are two main obstacles to truly individualize treatment with ICIs. One of the most severe and life-threatening adverse events is colitis induced colonic perforation, estimated to occur in 1.0 to 1.5% of patients treated with ICIs. An important question to address is, under what circumstances is it appropriate to reinitiate ICI treatment post-bowel perforation? CASE PRESENTATION The patient is a 62-year-old woman, who presented with stage IV lung cancer. Immunohistochemical staining indicated that 80% of the patient's tumor cells expressed PD-L1. The patient was started on a three-week cycle of pembrolizumab. Subsequent reducing in tumor burden was observed within ten weeks. Initially, pembrolizumab was tolerated fairly well, with the exception of immunotherapy related hypothyroidism. However, the patient experienced a second, more serious immune-related adverse event (irAE), in the form of enteritis, which led to small bowel perforation and necessitated exploratory laparotomy. The concerning part of the small bowel was resected, and a primary anastomosis was created. Based on the pathological and surgical findings, the patient was diagnosed with pembrolizumab-associated small bowel perforation. The patient recovered well from surgery and, considering the patient's remarkable response to treatment, a collective decision was made to reinitiate pembrolizumab on post-operative day twenty-eight. The patient is continuing her immunotherapy with ongoing partial response and is able to continue her full-time job. CONCLUSIONS This case report highlights the challenges of identifying patients likely to respond to ICIs and those that are likely to experience irAEs and it discusses the impressive work that has been done to start to address these challenges. Lastly, the topic of reinitiating pembrolizumab treatment even after colonic perforation is discussed.
Collapse
Affiliation(s)
- Tim N Beck
- Molecular and Cell Biology and Genetics, Drexel University College of Medicine, Philadelphia, PA, 19129, USA. .,Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA. .,Department of General Surgery, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Alexander E Kudinov
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Essel Dulaimi
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Yanis Boumber
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA. .,Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA. .,Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
| |
Collapse
|
37
|
Druzhkova I, Ignatova N, Prodanets N, Kiselev N, Zhukov I, Shirmanova M, Zagainov V, Zagaynova E. E-Cadherin in Colorectal Cancer: Relation to Chemosensitivity. Clin Colorectal Cancer 2019; 18:e74-e86. [PMID: 30415989 DOI: 10.1016/j.clcc.2018.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 04/13/2018] [Accepted: 10/09/2018] [Indexed: 02/07/2023]
|
38
|
Sansone A, Kliesch S, Isidori AM, Schlatt S. AMH and INSL3 in testicular and extragonadal pathophysiology: what do we know? Andrology 2019; 7:131-138. [DOI: 10.1111/andr.12597] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/09/2019] [Accepted: 01/22/2019] [Indexed: 12/18/2022]
Affiliation(s)
- A. Sansone
- Center of Reproductive Medicine and Andrology Department of Clinical and Surgical Andrology Institute of Reproductive and Regenerative Biology Münster Germany
- Department of Experimental Medicine Section of Medical Pathophysiology Food Science and Endocrinology – Sapienza University of Rome Rome Italy
| | - S. Kliesch
- Center of Reproductive Medicine and Andrology Department of Clinical and Surgical Andrology Institute of Reproductive and Regenerative Biology Münster Germany
| | - A. M. Isidori
- Department of Experimental Medicine Section of Medical Pathophysiology Food Science and Endocrinology – Sapienza University of Rome Rome Italy
| | - S. Schlatt
- Center of Reproductive Medicine and Andrology Department of Clinical and Surgical Andrology Institute of Reproductive and Regenerative Biology Münster Germany
| |
Collapse
|
39
|
Pellatt AJ, Mullany LE, Herrick JS, Sakoda LC, Wolff RK, Samowitz WS, Slattery ML. The TGFβ-signaling pathway and colorectal cancer: associations between dysregulated genes and miRNAs. J Transl Med 2018; 16:191. [PMID: 29986714 PMCID: PMC6038278 DOI: 10.1186/s12967-018-1566-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/29/2018] [Indexed: 12/19/2022] Open
Abstract
Background The TGFβ-signaling pathway plays an important role in the pathogenesis of colorectal cancer (CRC). Loss of function of several genes within this pathway, such as bone morphogenetic proteins (BMPs) have been seen as key events in CRC progression. Methods In this study we comprehensively evaluate differential gene expression (RNASeq) of 81 genes in the TGFβ-signaling pathway and evaluate how dysregulated genes are associated with miRNA expression (Agilent Human miRNA Microarray V19.0). We utilize paired carcinoma and normal tissue from 217 CRC cases. We evaluate the associations between differentially expressed genes and miRNAs and sex, age, disease stage, and survival months. Results Thirteen genes were significantly downregulated and 14 were significantly upregulated after considering fold change (FC) of > 1.50 or < 0.67 and multiple comparison adjustment. Bone morphogenetic protein genes BMP5, BMP6, and BMP2 and growth differentiation factor GDF7 were downregulated. BMP4, BMP7, INHBA (Inhibin beta A), TGFBR1, TGFB2, TGIF1, TGIF2, and TFDP1 were upregulated. In general, genes with the greatest dysregulation, such as BMP5 (FC 0.17, BMP6 (FC 0.25), BMP2 (FC 0.32), CDKN2B (FC 0.32), MYC (FC 3.70), BMP7 (FC 4.17), and INHBA (FC 9.34) showed dysregulation in the majority of the population (84.3, 77.4, 81.1, 80.2, 82.0, 51.2, and 75.1% respectively). Four genes, TGFBR2, ID4, ID1, and PITX2, were un-associated or slightly upregulated in microsatellite-stable (MSS) tumors while downregulated in microsatellite-unstable (MSI) tumors. Eight dysregulated genes were associated with miRNA differential expression. E2F5 and THBS1 were associated with one or two miRNAs; RBL1, TGFBR1, TGIF2, and INHBA were associated with seven or more miRNAs with multiple seed-region matches. Evaluation of the joint effects of mRNA:miRNA identified interactions that were stronger in more advanced disease stages and varied by survival months. Conclusion These data support an interaction between miRNAs and genes in the TGFβ-signaling pathway in association with CRC risk. These interactions are associated with unique clinical characteristics that may provide targets for further investigations. Electronic supplementary material The online version of this article (10.1186/s12967-018-1566-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Lila E Mullany
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT, 84108, USA
| | - Jennifer S Herrick
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT, 84108, USA
| | - Lori C Sakoda
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Roger K Wolff
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT, 84108, USA
| | - Wade S Samowitz
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Martha L Slattery
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT, 84108, USA.
| |
Collapse
|
40
|
Taximaimaiti R, Abudujilile D, Maihemuti M, Abuliken X, Abudulimu H. Expression of AMHR2 and C-KIT in cervical lesions in Uyghur Women of Xinjiang, China. Medicine (Baltimore) 2018; 97:e10793. [PMID: 29851788 PMCID: PMC6393133 DOI: 10.1097/md.0000000000010793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Cervical cancer is one of the most common malignant tumors in women. Anti-Müllerian hormone receptor 2 (AMHR2) and C-Kit were two members of protein kinase which were reported increased in some cancers like ovarian carcinoma and breast cancer. The present study aimed to assess the expression of AMHR2 and c-Kit in cervical cancer of different differentiated degrees as well as in cervicitis sections. METHODS All the lesions were collected randomly during clinical observations in hospitals located in Xinjiang, China. Polymerase chain reaction (PCR) and immunohistochemical staining were used to detect AMHR2 and c-Kit expression in cervical samples from women who had been infected with human papilloma virus (HPV)16. The expression rate was compared between cervical cancer of well, moderately and poorly differentiated and cervicitis. RESULTS The average age of the patients was 45 years; ranged from 23 to 80. For AMHR2, all 17 cervicitis samples ranged from (++) to (++++), while cervical cancer showed 11 (+), 9 (++), 15 (+++),9 (++++), and 8 (-), which showed AMHR2 expression was lessen with the poorer of differentiation degree of cervical cancer (P < .05). For c-Kit, 18 cervicitis samples mainly expressed as (-) with none showed (+++) or (++++), while cervical cancer samples showed 7 (-), 6 (+), 1 (++), 2 (+++), and 8 (++++), which indicated c-Kit's expression increased with the reduction of cervical cancer's differentiation degree (P < .05). CONCLUSION AMHR2 might have some correlation with self defense of our body, while c-Kit might link with the potential invasive capacity of cervical cancer.
Collapse
Affiliation(s)
- Reyisha Taximaimaiti
- Seven-year Clinical Medicine of Class 2, Xinjiang Medical University, Urumqi, PR China
| | - Dilinuer Abudujilile
- Seven-year Clinical Medicine of Class 2, Xinjiang Medical University, Urumqi, PR China
| | - Muzhapaer Maihemuti
- Seven-year Clinical Medicine of Class 2, Xinjiang Medical University, Urumqi, PR China
| | - Xiekelai Abuliken
- Seven-year Clinical Medicine of Class 2, Xinjiang Medical University, Urumqi, PR China
| | | |
Collapse
|
41
|
Rong B, Yang S. Molecular mechanism and targeted therapy of Hsp90 involved in lung cancer: New discoveries and developments (Review). Int J Oncol 2017; 52:321-336. [PMID: 29207057 DOI: 10.3892/ijo.2017.4214] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 06/13/2017] [Indexed: 11/05/2022] Open
Abstract
The exploration of the molecular mechanisms and signaling pathways on lung cancer is very important for developing new strategies of diagnosis and treatment to this disease, such as finding valuable lung cancer markers and molecularly targeted therapies. Previously, a number of studies disclose that heat shock protein 90 (Hsp90) is upregulated in cancer cells, tissues and serum of lung cancer patients, and its upregulation intimately correlates with the occurrence, development and outcome of lung cancer. On the contrary, inhibition of Hsp90 can suppress cell proliferation, motility and metastasis of lung cancer and promote apoptosis of lung cancer cells via complex signaling pathways. In addition, a series of Hsp90 inhibitors have been investigated as effective molecular targeted therapy tactics fighting against lung cancer. This review, systematically summarizes the role of Hsp90 in lung cancer, the molecular mechanisms and development of anti-Hsp90 treatment in lung cancer.
Collapse
Affiliation(s)
- Biaoxue Rong
- Department of Oncology, First Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, P.R. China
| | - Shuanying Yang
- Department of Respiratory Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| |
Collapse
|
42
|
Bougherara H, Némati F, Nicolas A, Massonnet G, Pugnière M, Ngô C, Le Frère-Belda MA, Leary A, Alexandre J, Meseure D, Barret JM, Navarro-Teulon I, Pèlegrin A, Roman-Roman S, Prost JF, Donnadieu E, Decaudin D. The humanized anti-human AMHRII mAb 3C23K exerts an anti-tumor activity against human ovarian cancer through tumor-associated macrophages. Oncotarget 2017; 8:99950-99965. [PMID: 29245952 PMCID: PMC5725143 DOI: 10.18632/oncotarget.21556] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 05/31/2017] [Indexed: 02/05/2023] Open
Abstract
Müllerian inhibiting substance, also called anti-Müllerian hormone (AMH), inhibits proliferation and induces apoptosis of AMH type II receptor-positive tumor cells, such as human ovarian cancers (OCs). On this basis, a humanized glyco-engineered monoclonal antibody (3C23K) has been developed. The aim of this study was therefore to experimentally confirm the therapeutic potential of 3C23K in human OCs. We first determined by immunofluorescence, immunohistochemistry and cytofluorometry analyses the expression of AMHRII in patient’s tumors and found that a majority (60 to 80% depending on the detection technique) of OCs were positive for this marker. We then provided evidence that the tumor stroma of OC is enriched in tumor-associated macrophages and that these cells are responsible for 3C23K-induced killing of tumor cells through ADCP and ADCC mechanisms. In addition, we showed that 3C23K reduced macrophages induced-T cells immunosuppression. Finally, we evaluated the therapeutic efficacy of 3C23K alone and in combination with a carboplatin-paclitaxel chemotherapy in a panel of OC Patient-Derived Xenografts. In those experiments, we showed that 3C23K significantly increased the proportion and the quality of chemotherapy-based in vivo responses. Altogether, our data support the potential interest of AMHRII targeting in human ovarian cancers and the evaluation of 3C23K in further clinical trials.
Collapse
Affiliation(s)
- Houcine Bougherara
- Inserm, U1016, Institut Cochin, Paris, France.,Cnrs, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Fariba Némati
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, Paris, France
| | - André Nicolas
- Department of Tumor Biology, Institut Curie, Paris, France
| | - Gérald Massonnet
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, Paris, France
| | - Martine Pugnière
- INSERM U896, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
| | - Charlotte Ngô
- Department of Gynaecological and Oncological Surgery, Hôpital Européen Georges Pompidou, Université Paris Descartes, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marie-Aude Le Frère-Belda
- Department of Pathology, Hôpital Européen Georges Pompidou, Université Paris Descartes, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Jérôme Alexandre
- Inserm, U1016, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Medical Oncology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Didier Meseure
- Department of Tumor Biology, Institut Curie, Paris, France
| | | | | | - André Pèlegrin
- INSERM U896, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
| | - Sergio Roman-Roman
- Department of Translational Research, Institut Curie, PSL University, Paris, France
| | | | - Emmanuel Donnadieu
- Inserm, U1016, Institut Cochin, Paris, France.,Cnrs, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Didier Decaudin
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, Paris, France.,Department of Medical Oncology, Institut Curie, Paris, France
| |
Collapse
|
43
|
Juratli TA, Qin N, Cahill DP, Filbin MG. Molecular pathogenesis and therapeutic implications in pediatric high-grade gliomas. Pharmacol Ther 2017; 182:70-79. [PMID: 28830841 DOI: 10.1016/j.pharmthera.2017.08.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
High-grade gliomas (HGG) are the most common malignant brain tumors in the pediatric population and account for a large subset of all pediatric central nervous system neoplasms. The management of pediatric HGG continues to be challenging, with poor outcome in many cases despite aggressive treatments. Consequently, parallel research efforts have been focused on identifying the underlying genetic and biological basis of pediatric HGG in order to more clearly define prognostic subgroups for treatment stratification as well as identify new treatment targets. These cutting-edge advances have revolutionized pediatric neuro-oncology and have revealed novel oncogenic vulnerabilities that are being therapeutically leveraged. Promising treatments - including pathway-targeting small molecules as well as epigenetic therapy - are being evaluated in clinical trials, and recent genomic discoveries in rare glioma subgroups have led to the identification of additional new potentially-actionable alterations. This review summarizes the current state of knowledge about the molecular characterization of pediatric HGG in correlation to the revised World Health Organization (WHO) classification, as well as provides an overview of some targeted treatment approaches in the modern clinical management of high-grade gliomas.
Collapse
Affiliation(s)
- Tareq A Juratli
- Translational Neuro-Oncology Laboratory, Department of Neurosurgery, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA; Department of Neurosurgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Germany.
| | - Nan Qin
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany; Division of Pediatric Neuro-Oncogenomics, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) - partner site Essen/Düsseldorf, Düsseldorf, Germany; Institute of Neuropathology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Daniel P Cahill
- Translational Neuro-Oncology Laboratory, Department of Neurosurgery, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mariella G Filbin
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Cancer and Blood Disorders Center, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02215, USA.
| |
Collapse
|
44
|
Serum variations of anti-mullerian hormone and total testosterone with aging in healthy adult Iranian men: A population-based study. PLoS One 2017; 12:e0179634. [PMID: 28715487 PMCID: PMC5513413 DOI: 10.1371/journal.pone.0179634] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/01/2017] [Indexed: 01/30/2023] Open
Abstract
Background Literature proves anti-mullerian hormone (AMH) and total testosterone (TT) as two important reproductive hormones in male development, however evidence regarding age variations of these hormones is lacking. Aims To estimate the normal serum AMH values and to assess the age-specific TT levels in men aged 30–70, we conducted the present population-based study. Methods A total of 831 healthy eligible men, aged 30–70 years, were recruited from Tehran Lipid and Glucose study cohort. Centiles for AMH were estimated according to the exponential normal 3-parameter model. The parametric method of Royston available in general software was applied for the first time to estimate the age-specific AMH and TT percentiles of 5th, 10th, 25th, 50th, 75th, 90th and 95th. Results Mean AMH level was 6.93, ranging from 0.1 to 40.1 ng/ml. Serum AMH concentrations followed a steady reduction with increasing age. Mean TT level was 4.8, ranging from 0.44 to 11.4 ng/ml. Discussion A measurable serum concentrations of AMH in healthy males throughout lifespan with variations, based on age, confirming a slight age-related AMH decline. Fractional polynomial (FP) regression models revealed that the mean and standard deviation (SD) of the TT were not associated with age, so the percentiles estimated were not age-specific. Conclusion We presented a nomogram of age-specific AMH values in a healthy cohort of Iranian men. This finding might have clinical importance in dealing hormonal disorders in men.
Collapse
|
45
|
Deneka AY, Haber L, Kopp MC, Gaponova AV, Nikonova AS, Golemis EA. Tumor-targeted SN38 inhibits growth of early stage non-small cell lung cancer (NSCLC) in a KRas/p53 transgenic mouse model. PLoS One 2017; 12:e0176747. [PMID: 28453558 PMCID: PMC5409145 DOI: 10.1371/journal.pone.0176747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/14/2017] [Indexed: 11/19/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer death worldwide, with a 5-year survival of only ~16%. Potential strategies to address NSCLC mortality include improvements in early detection and prevention, and development of new therapies suitable for use in patients with early and late stage diagnoses. Controlling the growth of early stage tumors could yield significant clinical benefits for patients with comorbidities that make them poor candidates for surgery: however, many drugs that limit cancer growth are not useful in the setting of long-term use or in comorbid patients, because of associated toxicities. In this study, we explored the use of a recently described small molecule agent, STA-8666, as a potential agent for controlling early stage tumor growth. STA-8666 uses a cleavable linker to merge a tumor-targeting moiety that binds heat shock protein 90 (HSP90) with the cytotoxic chemical SN38, and has been shown to have high efficacy and low toxicity, associated with efficient tumor targeting, in preclinical studies using patient-derived and other xenograft models for pancreatic, bladder, and small cell lung cancer. Using a genetically engineered model of NSCLC arising from induced mutation of KRas and knockout of Trp53, we continuously dosed mice with STA-8666 from immediately after tumor induction for 15 weeks. STA-8666 significantly slowed the rate of tumor growth, and was well tolerated over this extended dosing period. STA-8666 induced DNA damage and apoptosis, and reduced proliferation and phosphorylation of the proliferation-associated protein ERK1/2, selectively in tumor tissue. In contrast, STA-8666 did not affect tumor features, such as degree of vimentin staining, associated with epithelial-mesenchymal transition (EMT), or downregulate tumor expression of HSP90. These data suggest STA-8666 and other similar targeted compounds may be useful additions to control the growth of early stage NSCLC in patient populations.
Collapse
Affiliation(s)
- Alexander Y. Deneka
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
- Department of Biochemistry, Kazan Federal University, Kazan, Russia
- * E-mail: (EG); (AD)
| | - Leora Haber
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Meghan C. Kopp
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
- Program in Molecular and Cell Biology and Genetics, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Anna V. Gaponova
- Laboratory of Genome Engineering, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Immanuel Kant Baltic Federal University, Konigsberg, Russia
| | - Anna S. Nikonova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Erica A. Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
- * E-mail: (EG); (AD)
| |
Collapse
|