1
|
Kilari T, Suresh AS, Begum RF, Singh A, Venkkatesh P, Vellapandian C. Effect of Per and Poly-Fluoroalkyl Substances on Pregnancy and Child Development. Curr Pediatr Rev 2025; 21:142-153. [PMID: 38213179 DOI: 10.2174/0115733963267526231120110100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/18/2023] [Accepted: 11/02/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Childhood obesity is significantly influenced by maternal exposure to Per and Poly-Fluoroalkyl Substances (PFAS) during pregnancy. PFAS exposure occurs through the Peroxisome Proliferator-Activated Receptor (PPAR-γ) receptor, leading to increased fat deposition and profound health effects in child growth and development. Despite ongoing investigations, the relationship between maternal serum PFAS concentration and child obesity requires further exploration. OBJECTIVE This study aimed to review the possible effects of Per and poly-fluoroalkyl substances exposure and their mechanism in overweight/obese children from pregnant ladies. METHODS A detailed literature survey was conducted using online databases, including Science Direct, Google Scholar, Scopus, Cochrane, and PubMed. The study focused on the diverse effects of PFAS on maternal and child health, with particular emphasis on neurological complications. RESULTS Child growth development depends upon breastfeeding and placenta health, which is disrupted by PFAS exposure, ultimately destroying the body mass index of the child. Neurotoxicity testing utilized the SH-SY5Y human-derived cell line as an in vitro model, revealing PFAS-induced increases in adipocyte number, reduced cell size, altered lipid conglomeration, increased adiposity, and changes in liver function. in vivo studies in mice and human cell lines indicated PPAR-γ and ER-α activation, leading to adiposity and weight gain through Estrogen signaling and Lipid metabolism. PFAS concentrations positively correlated in maternal sera, analyzed by liquid chromatography/quadrupole mass spectrometry. CONCLUSION PFAS, with a long half-life of 3.5-8.5 years, is commonly found in the serum of pregnant women, crossing the placenta barrier. This exposure disrupts placental homeostasis, negatively impacting mechanisms of action and potentially leading to deterioration in pregnancy and child health. Further research is needed to comprehensively understand the complex interplay between PFAS exposure and its implications for maternal and child well-being.
Collapse
Affiliation(s)
- Thanuja Kilari
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur-603209, Tamil Nadu, India
| | - Ankul Singh Suresh
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur-603209, Tamil Nadu, India
| | - Rukaiah F Begum
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur-603209, Tamil Nadu, India
| | - Anuragh Singh
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur-603209, Tamil Nadu, India
| | - Pravin Venkkatesh
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur-603209, Tamil Nadu, India
| | - Chitra Vellapandian
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur-603209, Tamil Nadu, India
| |
Collapse
|
2
|
Albarracín-Navas L, Almonte-Becerril M, Guerrero E, Rivadeneira J, Telechea-Fernández M, Guzmán E, Calderón F, Hernández-Leal MJ, Otzen T, Manterola C, Duque G, Riffo-Campos ÁL. Differential Protein-Coding Gene Expression Profile in Patients with Prostate Cancer. Biomedicines 2024; 12:2509. [PMID: 39595075 PMCID: PMC11592252 DOI: 10.3390/biomedicines12112509] [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: 07/17/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Prostate cancer is the second most common neoplasm in men, with projections estimating over one million new cases by 2045. Differentially expressed genes can significantly enhance the diagnosis, treatment, monitoring, and prognosis of this disease. PURPOSE to systematically review and analyze validated differentially expressed mRNAs in prostate cancer patients to propose a robust molecular profile for clinical diagnostics. METHODS A systematic review was conducted following PRISMA guidelines, searching literature databases for mRNAs with validated differential expression in adult prostate cancer patients. Identified mRNAs were analyzed using STRING, Cytoscape, and DrugBank to explore protein-protein interactions and potential drug targets. RESULTS A total of 5003 participants from Europe, Asia, America, and Oceania were included, and 144 mRNAs (p < 0.05) were reported across 75 primary articles, predominantly validated using RT-qPCR with tissue samples. Among these, at least 36 mRNAs were identified as targets for cancer-related drugs. Enrichment analysis revealed the top pathways were associated with cancer, including specific prostate cancer terms. Key nodes emerged as hubs in the protein-protein interaction network. CONCLUSION Based on our comprehensive in silico analysis of validated differentially expressed mRNAs, we propose a molecular profile of twenty-five mRNAs with significant potential for clinical diagnosis of prostate cancer. These findings offer a valuable foundation for developing precision oncology strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Lorena Albarracín-Navas
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Comprehensive Medical Services (SERMEDIC), Cuenca 010111, Ecuador
| | - Maylin Almonte-Becerril
- Executive Direction of Research and Advanced Studies, Universidad de la Salud, Mexico City 01210, Mexico;
| | - Enmanuel Guerrero
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Comprehensive Medical Services (SERMEDIC), Cuenca 010111, Ecuador
- Faculty of Medicine, Universidad de Cuenca, Cuenca 010201, Ecuador
- SOLCA Cancer Institute, Cuenca 010111, Ecuador
| | - Josue Rivadeneira
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Zero Biomedical Research, Quito 170103, Ecuador
| | | | - Elizabeth Guzmán
- Comprehensive Medical Services (SERMEDIC), Cuenca 010111, Ecuador
| | | | - María José Hernández-Leal
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- University of Navarra, School of Nursing, Department of Community, Maternity and Pediatric Nursing, 31006 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Tamara Otzen
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Center for Cancer Prevention and Control (CECAN), Santiago 8380453, Chile
| | - Carlos Manterola
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Center for Cancer Prevention and Control (CECAN), Santiago 8380453, Chile
- Universidad de La Frontera, Center for Morphological and Surgical Studies (CEMyQ), Temuco 4811230, Chile
| | - Galo Duque
- Faculty of Medicine, Universidad del Azuay, Cuenca 010107, Ecuador;
| | - Ángela L. Riffo-Campos
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Center for Cancer Prevention and Control (CECAN), Santiago 8380453, Chile
| |
Collapse
|
3
|
Kim S, Chaudhary PK, Kim S. Molecular and Genetics Perspectives on Primary Adrenocortical Hyperfunction Disorders. Int J Mol Sci 2024; 25:11341. [PMID: 39518893 PMCID: PMC11545009 DOI: 10.3390/ijms252111341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Adrenocortical disorders encompass a broad spectrum of conditions ranging from benign hyperplasia to malignant tumors, significantly disrupting hormone balance and causing a variety of clinical manifestations. By leveraging next-generation sequencing and in silico analyses, recent studies have uncovered the genetic and molecular pathways implicated in these transitions. In this review, we explored the molecular and genetic alterations in adrenocortical disorders, with a particular focus on the transitions from normal adrenal function to hyperfunction. The insights gained are intended to enhance diagnostic and therapeutic strategies, offering up-to-date knowledge for managing these complex conditions effectively.
Collapse
Affiliation(s)
| | | | - Soochong Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (S.K.); (P.K.C.)
| |
Collapse
|
4
|
Rey RA. Steroid receptors in the testis: implications in the physiology of prenatal and postnatal development and translation to clinical application. Histol Histopathol 2023; 38:373-389. [PMID: 36218320 DOI: 10.14670/hh-18-533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023]
Abstract
The testes are the main source of sex steroids in the male, especially androgens and to a lesser extent estrogens. In target cells, steroid hormones typically signal after binding to intracellular receptors, which act as transcription factors. Androgens and estrogens have ubiquitous functions in peripheral organs, but also have paracrine actions within the gonads where they are far more concentrated. The levels of steroid production by the testes vary throughout fetal and postnatal development: they are high in intrauterine life and in the first months after birth, then they decline and are almost undetectable in childhood and increase again during puberty to attain adult levels. The expression of the androgen and estrogen receptors also depict specific ontogenies in the various testicular cell types. The combination of intratesticular steroid concentration with the pattern of expression of the steroid hormone receptors defines androgen and estrogen action on Sertoli, germ and Leydig cells. Here, we review the ontogeny of expression of the androgen and estrogen receptors in the testis, its impact on testicular physiology during prenatal and postnatal development, as well as its implication on the pathophysiology of different disorders affecting gonadal function throughout life.
Collapse
Affiliation(s)
- Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina.
- Unidad de Medicina Traslacional, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
- Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
5
|
Muacevic A, Adler JR, Eid FA, Alaswad HA, Ali WM, Aladraj FJ. Effects of Hormones and Endocrine Disorders on Hair Growth. Cureus 2022; 14:e32726. [PMID: 36578854 PMCID: PMC9788837 DOI: 10.7759/cureus.32726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Hormones have a close association with hair growth; thus, they have a big impact on the hair cycle and hair follicle structure. Many hormones control hair growth, cycle, and density. Hair abnormalities are frequent in therapeutic practice, and they can cause severe emotional discomfort depending on societal and ethnic standards. As a result, disorders that impact the endocrine system can induce a variety of physiological hair growth and cycling alterations. Hirsutism and patterned hair loss have a significant impact on human personality. These illnesses necessitate a comprehensive approach to diagnosis and treatment. The hormonal impact on hair growth and the association of different endocrine disorders with hair changes are briefly discussed here.
Collapse
|
6
|
Abstract
Androgens are essential sex steroid hormones for both sexes. Testosterone (T) is the predominant androgen in males, while in adult females, T concentrations are about 15-fold lower and androgen precursors are converted to estrogens. T is produced primarily in testicular Leydig cells in men, while in women precursors are biosynthesised in the adrenal cortex and ovaries and converted into T in the periphery. The biosynthesis of T occurs via a series of enzymatic reactions in steroidogenic organs. Notably, the more potent androgen, dihydrotestosterone, may be synthesized from T in the classic pathway, however, alternate metabolic pathways also exist. The classic action of androgens on target organs is mediated through the androgen receptor, which regulates nuclear receptor gene transcription. However, the androgen-androgen receptor complex may also interact directly with membrane proteins or signaling molecules to exert more rapid effects. This review summarizes the current knowledge of androgen biosynthesis, mechanisms of action and endocrine effects in human biology, and relates these effects to respective human congenital and acquired disorders.
Collapse
Affiliation(s)
- Rawda Naamneh Elzenaty
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern University Hospital, University of Bern, Switzerland; Department of Biomedical Research, University of Bern, Switzerland; Graduate School of Cellular and Biomedical Sciences, University of Bern, Switzerland.
| | - Therina du Toit
- Department of Biomedical Research, University of Bern, Switzerland.
| | - Christa E Flück
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern University Hospital, University of Bern, Switzerland; Department of Biomedical Research, University of Bern, Switzerland.
| |
Collapse
|
7
|
Shi R, Li S, Liu P, Guo L, Gong S, Wan Y. Effects of testosterone on skin structure and factors related to androgen conversion and binding in Hetian sheep. Trop Anim Health Prod 2022; 54:218. [PMID: 35759149 DOI: 10.1007/s11250-022-03216-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 06/18/2022] [Indexed: 12/06/2022]
Abstract
The effects of androgens on human skin mainly include the regulation of growth and differentiation of hair follicles and sebaceous glands. Androgens may have some physiological roles in sheep skin that are similar to those of humans, but further confirmation is needed. Therefore, Hetian sheep were chosen in this study as an animal model to explore the effects of testosterone on skin structure and factors related to androgen conversion and binding in Hetian sheep. The sheep were treated with different concentrations of testosterone for 42 days. Skin tissue sections were prepared and then subjected to hematoxylin-eosin, Sacpic, Masson's trichrome, and Oil Red O staining to observe changes in skin morphology. Changes in the content of blood-related factors were also detected using ELISA kits. The skin tissue distribution of androgen receptor was explored by immunohistochemistry and immunofluorescence assays. The results showed that testosterone significantly increases the sebaceous gland area and stimulates the formation of new sebaceous glands. Further exploration revealed that testosterone promotes the proliferation of sebaceous gland juvenile cells. However, testosterone was found to have no significant effect on hair follicle density and hair follicle structure. Testosterone increased dihydrotestosterone levels but decreased 5α-reductase 1 and 5α-reductase 2 levels. The androgen receptors were distributed in the hair follicles, sebaceous glands, and some major skin appendages of Hetian sheep. This study suggests that androgens can be effective in regulating sebum production in sheep. This study will help advance research efforts to further explore the molecular and cellular mechanisms by which androgens modify sheep follicles and sebaceous glands.
Collapse
Affiliation(s)
- Ruijun Shi
- Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, College of Life Science and Technology, Tarim University, Alar, Xinjiang, China
| | - Shuwei Li
- Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, College of Life Science and Technology, Tarim University, Alar, Xinjiang, China.
| | - Penggang Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Lili Guo
- Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, College of Life Science and Technology, Tarim University, Alar, Xinjiang, China
| | - Shujuan Gong
- Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, College of Life Science and Technology, Tarim University, Alar, Xinjiang, China
| | - Yu Wan
- Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, College of Life Science and Technology, Tarim University, Alar, Xinjiang, China
| |
Collapse
|
8
|
Mei Z, Yang T, Liu Y, Gao Y, Hou Z, Zhuang Q, He D, Zhang X, Tan Q, Zhu X, Qin Y, Chen X, Xu C, Bian C, Wang X, Wang C, Wu D, Huang S, Li Z. Management of prostate cancer by targeting 3βHSD1 after enzalutamide and abiraterone treatment. Cell Rep Med 2022; 3:100608. [PMID: 35584629 PMCID: PMC9133401 DOI: 10.1016/j.xcrm.2022.100608] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/27/2022] [Accepted: 03/25/2022] [Indexed: 12/19/2022]
Abstract
Novel strategies for prostate cancer therapy are required to overcome resistance to abiraterone and enzalutamide. Here, we show that increasing 3βHSD1 after abiraterone and enzalutamide treatment is essential for drug resistance, and biochanin A (BCA), as an inhibitor of 3βHSD1, overcomes drug resistance. 3βHSD1 activity increases in cell lines, biopsy samples, and patients after long-term treatment with enzalutamide or abiraterone. Enhanced steroidogenesis, mediated by 3βHSD1, is sufficient to impair enzalutamide function. In patients, accelerated abiraterone metabolism results in a decline of plasma abiraterone as disease progresses. BCA inhibits 3βHSD1 and suppresses prostate cancer development alone or together with abiraterone and enzalutamide. Daidzein, a BCA analog of dietary origin, is associated with higher plasma abiraterone concentrations and prevented prostate-specific antigen (PSA) increases in abiraterone-resistant patients. Overall, our results show that 3βHSD1 is a promising target to overcome drug resistance, and BCA suppresses disease progression as a 3βHSD1 inhibitor even after abiraterone and enzalutamide resistance.
Collapse
Affiliation(s)
- Zejie Mei
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Tao Yang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Ying Liu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yuanyuan Gao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zemin Hou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qian Zhuang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Dongyin He
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xuebin Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qilong Tan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xuyou Zhu
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yingyi Qin
- Department of Health Statistics, Second Military Medical University, No. 800 Xiangyin Road, Shanghai 200433, China
| | - Xi Chen
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Chengdang Xu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Cuidong Bian
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xinan Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Chenyang Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Denglong Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Shengsong Huang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Zhenfei Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| |
Collapse
|
9
|
Wisuitiprot V, Ingkaninan K, Chakkavittumrong P, Wisuitiprot W, Neungchamnong N, Chantakul R, Waranuch N. Effects of Acanthus ebracteatus Vahl. extract and verbascoside on human dermal papilla and murine macrophage. Sci Rep 2022; 12:1491. [PMID: 35087085 PMCID: PMC8795396 DOI: 10.1038/s41598-022-04966-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/04/2022] [Indexed: 12/22/2022] Open
Abstract
Androgenic alopecia is a common type of hair loss, usually caused by testosterone metabolism generating dihydrotestosterone and hair follicular micro-inflammation. These processes induce dermal papilla cells to undergo apoptosis. Currently approved effective medications for alopecia are Finasteride, an oral 5α-reductase inhibitor, Minoxidil, a topical hair growth promoter, and Diclofenac, an anti-inflammatory agent, all of which, however, have several adverse side effects. In our study, we showed the bioactivity of Acanthus ebracteatus Vahl. (AE) extract performed by 95% ethanol, and verbascoside (VB), a biomarker of AE extract. Both AE extract and VB were studied for their effects on dermal papilla cell viability and the cell cycle by using MTT assay and flow cytometry. The effect of an anti-inflammatory activity of AE extract and VB on IL-1β, NO, and TNF-α, released from LPS induced RAW 264.7 cells, and IL-1α and IL-6 released from irradiated dermal papilla cells were detected using ELISA technique. The preventive effect on dermal papilla cell apoptosis induced by testosterone was determined by MTT assay. In controlled in vitro assays it was found that AE extract and VB at various concentrations induced dermal papilla cell proliferation which was indicated by an increase in the number of cells in the S and G2/M phases of the cell cycle. AE extract at 250 µg/mL concentration or VB at 62.50 µg/mL concentration prevented cell apoptosis induced by testosterone at a statistically significant level. In addition, both AE extract and VB greatly inhibited the release of pro-inflammatory cytokines from RAW 264.7 and dermal papilla cells. The release of IL-1β, TNF-α, and NO from RAW 264.7 cells, as well as IL-1α and IL-6 from dermal papilla cells, was also diminished by AE extract 250 µg/mL and VB 125 µg/mL. Our results indicate that AE extract and VB are promising ingredients for anti-hair loss applications. However, further clinical study is necessary to evaluate the effectiveness of AE extract and VB as treatment for actual hair loss.
Collapse
Affiliation(s)
- Vanuchawan Wisuitiprot
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences and Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, 65000, Thailand
| | - Kornkanok Ingkaninan
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Panlop Chakkavittumrong
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Thammasat University, Khlong Luang, Pathumthani, 12121, Thailand
| | - Wudtichai Wisuitiprot
- Department of Thai Traditional Medicine, Sirindhorn College of Public Health, Phitsanulok, 65130, Thailand
| | - Nitra Neungchamnong
- Science Laboratory Centre, Faculty of Science, Naresuan University, Mueang, Phitsanulok, 65000, Thailand
| | - Ruttanaporn Chantakul
- Bioscreening Unit, Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Neti Waranuch
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences and Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok, 65000, Thailand. .,Cosmetics and Natural Products Research Center, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, 65000, Thailand.
| |
Collapse
|
10
|
Wang JM, Li ZF, Yang WX. What Does Androgen Receptor Signaling Pathway in Sertoli Cells During Normal Spermatogenesis Tell Us? Front Endocrinol (Lausanne) 2022; 13:838858. [PMID: 35282467 PMCID: PMC8908322 DOI: 10.3389/fendo.2022.838858] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/01/2022] [Indexed: 01/18/2023] Open
Abstract
Androgen receptor signaling pathway is necessary to complete spermatogenesis in testes. Difference between androgen binding location in Sertoli cell classifies androgen receptor signaling pathway into classical signaling pathway and non-classical signaling pathway. As the only somatic cell type in seminiferous tubule, Sertoli cells are under androgen receptor signaling pathway regulation via androgen receptor located in cytoplasm and plasma membrane. Androgen receptor signaling pathway is able to regulate biological processes in Sertoli cells as well as germ cells surrounded between Sertoli cells. Our review will summarize the major discoveries of androgen receptor signaling pathway in Sertoli cells and the paracrine action on germ cells. Androgen receptor signaling pathway regulates Sertoli cell proliferation and maturation, as well as maintain the integrity of blood-testis barrier formed between Sertoli cells. Also, Spermatogonia stem cells achieve a balance between self-renewal and differentiation under androgen receptor signaling regulation. Meiotic and post-meiotic processes including Sertoli cell - Spermatid attachment and Spermatid development are guaranteed by androgen receptor signaling until the final sperm release. This review also includes one disease related to androgen receptor signaling dysfunction named as androgen insensitivity syndrome. As a step further ahead, this review may be conducive to develop therapies which can cure impaired androgen receptor signaling in Sertoli cells.
Collapse
|
11
|
Eckhardt C, Sbiera I, Krebs M, Sbiera S, Spahn M, Kneitz B, Joniau S, Fassnacht M, Kübler H, Weigand I, Kroiss M. High expression of Sterol-O-Acyl transferase 1 (SOAT1), an enzyme involved in cholesterol metabolism, is associated with earlier biochemical recurrence in high risk prostate cancer. Prostate Cancer Prostatic Dis 2021; 25:484-490. [PMID: 34326474 PMCID: PMC9385470 DOI: 10.1038/s41391-021-00431-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 07/06/2021] [Accepted: 07/15/2021] [Indexed: 01/04/2023]
Abstract
Background Prostate cancer (PCa) is the most frequent cancer in men. The prognosis of PCa is heterogeneous with many clinically indolent tumors and rare highly aggressive cases. Reliable tissue markers of prognosis are lacking. Active cholesteryl ester synthesis has been associated with prostate cancer aggressiveness. Sterol-O-Acyl transferases (SOAT) 1 and 2 catalyze cholesterol esterification in humans. Objective To investigate the value of SOAT1 and SOAT2 tissue expression as prognostic markers in high risk PCa. Patients and methods Formalin-fixed paraffin-embedded tissue samples from 305 high risk PCa cases treated with radical prostatectomy were analyzed for SOAT1 and SOAT2 protein expression by semi-quantitative immunohistochemistry. The Kaplan–Meier method and Cox proportional hazards modeling were used to compare outcome. Main outcome measure Biochemical recurrence (BCR) free survival. Results SOAT1 expression was high in 73 (25%) and low in 219 (75%; not evaluable: 13) tumors. SOAT2 was highly expressed in 40 (14%) and at low levels in 249 (86%) samples (not evaluable: 16). By Kaplan–Meier analysis, we found significantly shorter median BCR free survival of 93 months (95% confidence interval 23.6–123.1) in patients with high SOAT1 vs. 134 months (112.6–220.2, Log-rank p < 0.001) with low SOAT1. SOAT2 expression was not significantly associated with BCR. After adjustment for age, preoperative PSA, tumor stage, Gleason score, resection status, lymph node involvement and year of surgery, high SOAT1 but not SOAT2 expression was associated with shorter BCR free survival with a hazard ratio of 2.40 (95% CI 1.57–3.68, p < 0.001). Time to clinical recurrence and overall survival were not significantly associated with SOAT1 and SOAT2 expression Conclusions SOAT1 expression is strongly associated with BCR free survival alone and after multivariable adjustment in high risk PCa. SOAT1 may serve as a histologic marker of prognosis and holds promise as a future treatment target.
Collapse
Affiliation(s)
- Carolin Eckhardt
- University Hospital Würzburg, Department of Internal Medicine I, Division of Endocrinology and Diabetology, Würzburg, Germany
| | - Iuliu Sbiera
- University Hospital Würzburg, Department of Internal Medicine I, Division of Endocrinology and Diabetology, Würzburg, Germany
| | - Markus Krebs
- University Hospital Würzburg, Department of Urology and Pediatric Urology, Würzburg, Germany.,University of Würzburg, Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Silviu Sbiera
- University Hospital Würzburg, Department of Internal Medicine I, Division of Endocrinology and Diabetology, Würzburg, Germany
| | - Martin Spahn
- Lindenhofspital, Bern, Switzerland.,Department of Urology, University Hospital Essen, Essen, Germany
| | - Burkhard Kneitz
- University Hospital Würzburg, Department of Urology and Pediatric Urology, Würzburg, Germany
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Martin Fassnacht
- University Hospital Würzburg, Department of Internal Medicine I, Division of Endocrinology and Diabetology, Würzburg, Germany.,University of Würzburg, Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Hubert Kübler
- University Hospital Würzburg, Department of Urology and Pediatric Urology, Würzburg, Germany.,University of Würzburg, Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Isabel Weigand
- University Hospital Würzburg, Department of Internal Medicine I, Division of Endocrinology and Diabetology, Würzburg, Germany.,Department of Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, München, Germany
| | - Matthias Kroiss
- University Hospital Würzburg, Department of Internal Medicine I, Division of Endocrinology and Diabetology, Würzburg, Germany. .,University of Würzburg, Comprehensive Cancer Center Mainfranken, Würzburg, Germany. .,Department of Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, München, Germany.
| |
Collapse
|
12
|
Schiewer MJ, Knudsen KE. Basic Science and Molecular Genetics of Prostate Cancer Aggressiveness. Urol Clin North Am 2021; 48:339-347. [PMID: 34210489 DOI: 10.1016/j.ucl.2021.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Androgen receptor function, tumor cell plasticity, loss of tumor suppressors, and defects in DNA repair genes affect aggressive features of prostate cancer. Prostate cancer development, progression, and aggressive behavior are often attributable to function of the androgen receptor. Tumor cell plasticity, neuroendocrine features, and loss of tumor suppressors lend aggressive behavior to prostate cancer cells. DNA repair defects have ramifications for prostate cancer cell behavior.
Collapse
Affiliation(s)
- Matthew J Schiewer
- Department of Urology, Urology Research Laboratory, Thomas Jefferson University, Sidney Kimmel Cancer Center, 233 South 10th Street BLSB 804, Philadelphia, PA 19107, USA; Department of Cancer Biology, Urology Research Laboratory, Thomas Jefferson University, Sidney Kimmel Cancer Center, 233 South 10th Street BLSB 804, Philadelphia, PA 19107, USA.
| | - Karen E Knudsen
- Department of Cancer Biology, Thomas Jefferson University, 233 South 10th Street BLSB 1050, Philadelphia, PA 19107, USA; Department of Urology, Thomas Jefferson University, 233 South 10th Street BLSB 1050, Philadelphia, PA 19107, USA; Department of Medical Oncology, Thomas Jefferson University, 233 South 10th Street BLSB 1050, Philadelphia, PA 19107, USA; Department of Radiation Oncology, Thomas Jefferson University, 233 South 10th Street BLSB 1050, Philadelphia, PA 19107, USA. https://twitter.com/SKCCDirector
| |
Collapse
|
13
|
Porter LH, Bakshi A, Pook D, Clark A, Clouston D, Kourambas J, Goode DL, Risbridger GP, Taylor RA, Lawrence MG. Androgen receptor enhancer amplification in matched patient-derived xenografts of primary and castrate-resistant prostate cancer. J Pathol 2021; 254:121-134. [PMID: 33620092 DOI: 10.1002/path.5652] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/30/2022]
Abstract
Amplifications of the androgen receptor (AR) occur in up to 80% of men with castration-resistant prostate cancer (CRPC). Recent studies highlighted that these amplifications not only span the AR gene but usually encompass a distal enhancer. This represents a newly recognised, non-coding mechanism of resistance to AR-directed therapies, including enzalutamide. To study disease progression before and after AR amplification, we used tumour samples from a castrate-sensitive primary tumour and castrate-resistant metastasis of the same patient. For subsequent functional and genomic studies, we established serially transplantable patient-derived xenografts (PDXs). Whole genome sequencing showed that alterations associated with poor prognosis, such as TP53 and PTEN loss, existed before androgen deprivation therapy, followed by co-amplification of the AR gene and enhancer after the development of metastatic CRPC. The PDX of the primary tumour, without the AR amplification, was sensitive to AR-directed treatments, including castration, enzalutamide, and apalutamide. The PDX of the metastasis, with the AR amplification, had higher AR and AR-V7 expression in castrate conditions, and was resistant to castration, apalutamide, and enzalutamide in vivo. Treatment with a BET inhibitor outperformed the AR-directed therapies for the metastasis, resulting in tumour regression for some, but not all, grafts. Therefore, this study provides novel matched PDXs to test potential treatments that target the overabundance of AR in tumours with AR enhancer amplifications. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Laura H Porter
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Andrew Bakshi
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - David Pook
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Medical Oncology, Monash Health, Clayton, VIC, Australia
| | - Ashlee Clark
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | | | - John Kourambas
- Department of Medicine, Monash Health, Casey Hospital, Berwick, VIC, Australia
| | -
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - David L Goode
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Gail P Risbridger
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Renea A Taylor
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Mitchell G Lawrence
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
14
|
Makwana V, Rudrawar S, Anoopkumar-Dukie S. Signalling transduction of O-GlcNAcylation and PI3K/AKT/mTOR-axis in prostate cancer. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166129. [PMID: 33744394 DOI: 10.1016/j.bbadis.2021.166129] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 02/26/2021] [Accepted: 03/14/2021] [Indexed: 12/23/2022]
Abstract
Hexosamine biosynthetic (HBP) and PI3K/AKT/mTOR pathways are found to predominate the proliferation and survival of prostate cancer cells. Both these pathways have their own specific intermediates to propagate the secondary signals in down-stream cascades and besides having their own structured network, also have shared interconnecting branches. These interconnections are either competitive or co-operative in nature depending on the microenvironmental conditions. Specifically, in prostate cancer HBP and mTOR pathways increases the expression and protein level of androgen receptor in order to support cancer cell proliferation, advancement and metastasis. Pharmacological inhibition of a single pathway is therefore insufficient to stop disease progression as the cancer cells manage to alter the signalling channel. This is one of the primary reasons for the therapeutic failure in prostate cancer and emergence of chemoresistance. Inhibition of these multiple pathways at their common junctures might prove to be of benefit in men suffering from an advanced disease state. Hence, a thorough understanding of these cellular intersecting points and their significance with respect to signal transduction mechanisms might assist in the rational designing of combinations for effective management of prostate cancer.
Collapse
Affiliation(s)
- Vivek Makwana
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, QLD 4222, Australia
| | - Santosh Rudrawar
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, QLD 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia; Quality Use of Medicines Network, Griffith University, Gold Coast, QLD 4222, Australia.
| | - Shailendra Anoopkumar-Dukie
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, QLD 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia; Quality Use of Medicines Network, Griffith University, Gold Coast, QLD 4222, Australia.
| |
Collapse
|
15
|
Lin C, Blessing AM, Pulliam TL, Shi Y, Wilkenfeld SR, Han JJ, Murray MM, Pham AH, Duong K, Brun SN, Shaw RJ, Ittmann MM, Frigo DE. Inhibition of CAMKK2 impairs autophagy and castration-resistant prostate cancer via suppression of AMPK-ULK1 signaling. Oncogene 2021; 40:1690-1705. [PMID: 33531625 PMCID: PMC7935762 DOI: 10.1038/s41388-021-01658-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 12/18/2020] [Accepted: 01/14/2021] [Indexed: 01/30/2023]
Abstract
Previous work has suggested androgen receptor (AR) signaling mediates prostate cancer progression in part through the modulation of autophagy. However, clinical trials testing autophagy inhibition using chloroquine derivatives in men with castration-resistant prostate cancer (CRPC) have yet to yield promising results, potentially due to the side effects of this class of compounds. We hypothesized that identification of the upstream activators of autophagy in prostate cancer could highlight alternative, context-dependent targets for blocking this important cellular process during disease progression. Here, we used molecular, genetic, and pharmacological approaches to elucidate an AR-mediated autophagy cascade involving Ca2+/calmodulin-dependent protein kinase kinase 2 (CAMKK2; a kinase with a restricted expression profile), 5'-AMP-activated protein kinase (AMPK), and Unc-51 like autophagy activating kinase 1 (ULK1), but independent of canonical mechanistic target of rapamycin (mTOR) activity. Increased CAMKK2-AMPK-ULK1 signaling correlated with disease progression in genetic mouse models and patient tumor samples. Importantly, CAMKK2 disruption impaired tumor growth and prolonged survival in multiple CRPC preclinical mouse models. Similarly, an inhibitor of AMPK-ULK1 blocked autophagy, cell growth, and colony formation in prostate cancer cells. Collectively, our findings converge to demonstrate that AR can co-opt the CAMKK2-AMPK-ULK1 signaling cascade to promote prostate cancer by increasing autophagy. Thus, this pathway may represent an alternative autophagic target in CRPC.
Collapse
Affiliation(s)
- Chenchu Lin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Alicia M Blessing
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Thomas L Pulliam
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Yan Shi
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
| | - Sandi R Wilkenfeld
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jenny J Han
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mollianne M Murray
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander H Pham
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
| | - Kevin Duong
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Sonja N Brun
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Reuben J Shaw
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michael M Ittmann
- Departments of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Houston, TX, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA.
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
16
|
Abstract
Puberty is characterized by major changes in the anatomy and function of reproductive organs. Androgen activity is low before puberty, but during pubertal development, the testes resume the production of androgens. Major physiological changes occur in the testicular cell compartments in response to the increase in intratesticular testosterone concentrations and androgen receptor expression. Androgen activity also impacts on the internal and external genitalia. In target cells, androgens signal through a classical and a nonclassical pathway. This review addresses the most recent advances in the knowledge of the role of androgen signaling in postnatal male sexual development, with a special emphasis on human puberty.
Collapse
Affiliation(s)
- Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, C1121ABG Buenos Aires, Argentina
| |
Collapse
|
17
|
Kamada S, Takeiwa T, Ikeda K, Horie-Inoue K, Inoue S. Long Non-coding RNAs Involved in Metabolic Alterations in Breast and Prostate Cancers. Front Oncol 2020; 10:593200. [PMID: 33123488 PMCID: PMC7573247 DOI: 10.3389/fonc.2020.593200] [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: 08/10/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Breast and prostate cancers are the most prevalent cancers in females and males, respectively. These cancers exhibit sex hormone dependence and thus, hormonal therapies are used to treat these cancers. However, acquired resistance to hormone therapies is a major clinical problem. In addition, certain portions of these cancers initially exhibit hormone-independence due to the absence of sex hormone receptors. Therefore, precise and profound understanding of the cancer pathophysiology is required to develop novel clinical strategies against breast and prostate cancers. Metabolic reprogramming is currently recognized as one of the hallmarks of cancer, as exemplified by the alteration of glucose metabolism, oxidative phosphorylation, and lipid metabolism. Dysregulation of metabolic enzymes and their regulators such as kinases, transcription factors, and other signaling molecules contributes to metabolic alteration in cancer. Moreover, accumulating lines of evidence reveal that long non-coding RNAs (lncRNAs) regulate cancer development and progression by modulating metabolism. Understanding the mechanism and function of lncRNAs associated with cancer-specific metabolic alteration will therefore provide new knowledge for cancer diagnosis and treatment. This review provides an overview of recent studies regarding the role of lncRNAs in metabolism in breast and prostate cancers, with a focus on both sex hormone-dependent and -independent pathways.
Collapse
Affiliation(s)
- Shuhei Kamada
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan.,Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshihiko Takeiwa
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan
| | - Kazuhiro Ikeda
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan
| | - Kuniko Horie-Inoue
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan
| | - Satoshi Inoue
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan.,Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
18
|
Abstract
The hair cycle and hair follicle structure are highly affected by various hormones. Androgens—such as testosterone (T); dihydrotestosterone (DHT); and their prohormones, dehydroepiandrosterone sulfate (DHEAS) and androstendione (A)—are the key factors in terminal hair growth. They act on sex-specific areas of the body, converting small, straight, fair vellus hairs into larger darker terminal hairs. They bind to intracellular androgen receptors in the dermal papilla cells of the hair follicle. The majority of hair follicles also require the intracellular enzyme 5-alpha reductase to convert testosterone into DHT. Apart from androgens, the role of other hormones is also currently being researched—e.g., estradiol can significantly alter the hair follicle growth and cycle by binding to estrogen receptors and influencing aromatase activity, which is responsible for converting androgen into estrogen (E2). Progesterone, at the level of the hair follicle, decreases the conversion of testosterone into DHT. The influence of prolactin (PRL) on hair growth has also been intensively investigated, and PRL and PRL receptors were detected in human scalp skin. Our review includes results from many analyses and provides a comprehensive up-to-date understanding of the subject of the effects of hormonal changes on the hair follicle.
Collapse
|
19
|
Katzenellenbogen JA. PET Imaging Agents (FES, FFNP, and FDHT) for Estrogen, Androgen, and Progesterone Receptors to Improve Management of Breast and Prostate Cancers by Functional Imaging. Cancers (Basel) 2020; 12:E2020. [PMID: 32718075 PMCID: PMC7465097 DOI: 10.3390/cancers12082020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/30/2020] [Accepted: 07/17/2020] [Indexed: 12/20/2022] Open
Abstract
Many breast and prostate cancers are driven by the action of steroid hormones on their cognate receptors in primary tumors and in metastases, and endocrine therapies that inhibit hormone production or block the action of these receptors provide clinical benefit to many but not all of these cancer patients. Because it is difficult to predict which individuals will be helped by endocrine therapies and which will not, positron emission tomography (PET) imaging of estrogen receptor (ER) and progesterone receptor (PgR) in breast cancer, and androgen receptor (AR) in prostate cancer can provide useful, often functional, information on the likelihood of endocrine therapy response in individual patients. This review covers our development of three PET imaging agents, 16α-[18F]fluoroestradiol (FES) for ER, 21-[18F]fluoro-furanyl-nor-progesterone (FFNP) for PgR, and 16β-[18F]fluoro-5α-dihydrotestosterone (FDHT) for AR, and the evolution of their clinical use. For these agents, the pathway from concept through development tracks with an emerging understanding of critical performance criteria that is needed for successful PET imaging of these low-abundance receptor targets. Progress in the ongoing evaluation of what they can add to the clinical management of breast and prostate cancers reflects our increased understanding of these diseases and of optimal strategies for predicting the success of clinical endocrine therapies.
Collapse
Affiliation(s)
- John A Katzenellenbogen
- Department of Chemistry and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
20
|
Manceau C, Mourey L, Pouessel D, Ploussard G. Abiraterone acetate in combination with prednisone in the treatment of prostate cancer: safety and efficacy. Expert Rev Anticancer Ther 2020; 20:629-638. [PMID: 32552120 DOI: 10.1080/14737140.2020.1785289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Metastatic prostate cancer is a life-threatening disease and an important public health concern with prevalence rates varying drastically between high- and low-income countries. Androgen-deprivation therapy alone has been the first-line treatment option for decades, temporarily controlling disease until invariable tumor regression. At the castration-resistant stage, metastatic disease becomes lethal. In recent years several new treatments, including second-generation hormone therapies, have proven to be life-prolonging in metastatic castration-resistant prostate cancer, and at an earlier hormone-sensitive stage. Abiraterone acetate in combination with prednisone was the first approved hormone therapy demonstrating survival benefit, and represents, to date, an alternative, or a second-line treatment after taxane-based chemotherapy, in addition to androgen-deprivation therapy, in hormone sensitive, and metastatic castration-resistant prostate cancer. AREA COVERED We performed a literature review of papers from 2012 to 2020 using PubMed, Web of Science, and Embase searching for the safety and efficacy of abiraterone acetate in prostate cancer management. Search results were limited to phase III-IV trials and post hoc analysis of Phase III trials evaluated Abiraterone acetate in the English language. EXPERT OPINION This literature review confirms the role of abiraterone acetate in the therapeutic landscape with well-proven safety and efficacy, demonstrated in trials and post-approval studies.
Collapse
Affiliation(s)
- Cécile Manceau
- Department of Urology, CHU-IUCT Oncopole , Toulouse, France
| | - Loic Mourey
- Department of Medical Oncology, Institut Claudius Régaud, IUCT Oncopole , Toulouse, France
| | - Damien Pouessel
- Department of Medical Oncology, Institut Claudius Régaud, IUCT Oncopole , Toulouse, France
| | | |
Collapse
|