1
|
Li F, Qi JJ, Li LX, Yan TF. MTHFR C677T、MTHFR A1298C、MTRR A66G and MTR A2756G polymorphisms and male infertility risk: a systematic review and meta-analysis. Reprod Biol Endocrinol 2024; 22:133. [PMID: 39478547 PMCID: PMC11523872 DOI: 10.1186/s12958-024-01306-7] [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] [Received: 06/21/2023] [Accepted: 10/23/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Epidemiological studies have reported that polymorphisms of folate-metabolizing genes have a significant impact on male infertility. However, the results of published studies have come to different conclusions. OBJECTIVE To determine an association between folate-metabolizing gene polymorphisms and the risk of male infertility. METHODS The meta-analysis was conducted according to the PRISMA 2020 statement. The protocol was registered with PROSPERO (CRD42023412251). Studies were searched from PubMed, Google Scholar, Embase, Scopus, and the Cochrane Library up to 24st October2023. Articles that satisfied the inclusion criteria were evaluated for their quality using the Newcastle-Ottawa Scale. Data were extracted from the eligible studies and were analyzed for pooled up odds ratio (OR) with 95% confidence interval (CI). Meta-analysis was conducted using STATA 12. RESULTS Forty-six case-control studies were included in the meta-analysis which comprised 20,639 participants. The pooled analysis revealed that the MTHFR C677T polymorphism was significantly associated with male infertility and abnormospermia.Three-fifths of the model showed there was a significant association between the MTR A2756G polymorphism and male infertility. Both MTHFR A1298C and MTRR A66G polymorphisms were not significantly associated with male fertility. Furthermore, subgroup analysis revealed a significant association between the MTHFR C677T polymorphism and male fertility in Asian countries. CONCLUSION This meta-analysis suggests that the MTHFR C677T and MTR A2756G polymorphisms may be a potential risk factor for male infertility.
Collapse
Affiliation(s)
- Feng Li
- Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, Hainan Province, 570311, China
| | - Ju-Ju Qi
- The First Hospital of Shijiazhuang, Shijiazhuang, Heibei Province, 050011, China
| | - Li-Xin Li
- The First Hospital of Shijiazhuang, Shijiazhuang, Heibei Province, 050011, China
| | - Teng-Fei Yan
- Baoding No.1, Central Hospital, Baoding, Hebei Province, 071000, China.
| |
Collapse
|
2
|
Weibring K, Lundberg FE, Cohn-Cedermark G, Rodriguez-Wallberg KA. Sperm Quality in 1252 Adolescents and Young Adults (AYAs) Undergoing Fertility Preservation Due to Cancer or Nonmalignant Diseases. J Adolesc Young Adult Oncol 2024. [PMID: 39069896 DOI: 10.1089/jayao.2024.0068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
Purpose: To investigate the quality of emergency-collected semen samples aimed at sperm cryopreservation provided by adolescents and young adults (AYAs) presenting with cancer or nonmalignant diseases. Methods: This is a prospective cohort study of postpubertal males referred for sperm cryopreservation who provided at least one semen sample for fertility preservation at the Reproductive Medicine Clinic of Karolinska University Hospital, Stockholm, Sweden, between January 2009 and January 2020. Sperm quality was assessed by total sperm count, concentration, and motility. Sperm quality by disease groups was compared with the reference population data of fertile men defined by the World Health Organization (WHO). Results: Among the 1252 patients who provided samples for cryopreservation, 1063 had cancer and 189 had nonmalignant diseases. The most common malignant indications included testicular cancers (n = 501) and Hodgkin lymphoma (n = 102). Among those with nonmalignant disease, 35% (n = 66) had testicular disease. Sperm quality was significantly lower in all groups of patients with cancer compared with the reference population. In total, azoospermia was found in 8% of the patients with cancer, in 9% of those with nonmalignant testicular disease, and in 3% of the remaining men with nonmalignant disease. Conclusion: Sperm quality in adult patients with cancer was significantly impaired compared with the WHO reference population standards for fertile men. For adolescent patients, standard reference values are lacking. AYAs wishing to preserve fertility should receive individualized counseling regarding sperm quality at the time of cryopreservation, and in selected cases, banking of additional samples should be recommended depending on the sperm quality parameters.
Collapse
Affiliation(s)
- Kristina Weibring
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Frida E Lundberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Gabriella Cohn-Cedermark
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Laboratory of Translational Fertility Preservation, Karolinska Institutet, Stockholm, Sweden
- Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
3
|
Galdon G, Zarandi NP, Deebel NA, Zhang S, Cornett O, Lyalin D, Pettenati MJ, Lue Y, Wang C, Swerdloff R, Shupe TD, Bishop C, Stogner K, Kogan SJ, Howards S, Atala A, Sadri-Ardekani H. In Vitro Generation of Haploid Germ Cells from Human XY and XXY Immature Testes in a 3D Organoid System. Bioengineering (Basel) 2024; 11:677. [PMID: 39061759 PMCID: PMC11274239 DOI: 10.3390/bioengineering11070677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
Increasing survival rates of children following cancer treatment have resulted in a significant population of adult survivors with the common side effect of infertility. Additionally, the availability of genetic testing has identified Klinefelter syndrome (classic 47,XXY) as the cause of future male infertility for a significant number of prepubertal patients. This study explores new spermatogonia stem cell (SSC)-based fertility therapies to meet the needs of these patients. Testicular cells were isolated from cryopreserved human testes tissue stored from XY and XXY prepubertal patients and propagated in a two-dimensional culture. Cells were then incorporated into a 3D human testicular organoid (HTO) system. During a 3-week culture period, HTOs maintained their structure, viability, and metabolic activity. Cell-specific PCR and flow cytometry markers identified undifferentiated spermatogonia, Sertoli, Leydig, and peritubular cells within the HTOs. Testosterone was produced by the HTOs both with and without hCG stimulation. Upregulation of postmeiotic germ cell markers was detected after 23 days in culture. Fluorescence in situ hybridization (FISH) of chromosomes X, Y, and 18 identified haploid cells in the in vitro differentiated HTOs. Thus, 3D HTOs were successfully generated from isolated immature human testicular cells from both euploid (XY) and Klinefelter (XXY) patients, supporting androgen production and germ cell differentiation in vitro.
Collapse
Affiliation(s)
- Guillermo Galdon
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Facultad de Medicina, Universidad de Barcelona, 08036 Barcelona, Spain
| | - Nima Pourhabibi Zarandi
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Internal Medicine, University of Pittsburgh Medical Center, Harrisburg, PA 17101, USA
| | - Nicholas A. Deebel
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Sue Zhang
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Olivia Cornett
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Dmitry Lyalin
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Department of Pathology, Molecular Diagnostics Division, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Mark J. Pettenati
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - YanHe Lue
- Division of Endocrinology, Department of Medicine, The Lundquist Institute, Harbor-University of California Los Angeles (UCLA) Medical Center, Los Angeles, CA 90502, USA
| | - Christina Wang
- Division of Endocrinology, Department of Medicine, The Lundquist Institute, Harbor-University of California Los Angeles (UCLA) Medical Center, Los Angeles, CA 90502, USA
| | - Ronald Swerdloff
- Division of Endocrinology, Department of Medicine, The Lundquist Institute, Harbor-University of California Los Angeles (UCLA) Medical Center, Los Angeles, CA 90502, USA
| | - Thomas D. Shupe
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Colin Bishop
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Kimberly Stogner
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Stanley J. Kogan
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Stuart Howards
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Hooman Sadri-Ardekani
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
4
|
Liu X, Wang Q, Sheng H, Liang X, Wang Z, Meng T, Li Y, Dong H, Zhu W, Yang J, Zhang Z, Jiang X, Zhang A, Liang Z, He X, Song C, Li F, Zhang X. Fertility preservation in males with cancer of trends, region development, and efficacy in mainland China from 16 regions Chinese sperm banks. J Assist Reprod Genet 2024; 41:1893-1906. [PMID: 38676843 PMCID: PMC11263528 DOI: 10.1007/s10815-024-03121-w] [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: 12/02/2023] [Accepted: 04/09/2024] [Indexed: 04/29/2024] Open
Abstract
PURPOSE Male cancer survivors experience confusion about fertility following cancer treatment. The aims of this study were to evaluate survivors' semen quality in different tumor type groups in China and to analyze the current situation and challenges of male cancer patients with sperm cryopreservation. METHODS This was a multicenter retrospective study of male patients with cancer who underwent sperm cryopreservation in 16 regions of the national sperm banks over an 11-year period from 2010 to 2020. RESULTS The number of male cancer patients with sperm cryopreservation showed an overall upward trend. The development of male cancer fertility preservation (FP) in the eastern, central, and western regions of Chinese displayed imbalance. There are seven tumor types for sperm preservation in the top incidence ten tumor types, including lymphoma, leukemia, nasopharyngeal carcinoma, sarcoma, thyroid cancer, and brain tumor. Moreover, nasopharyngeal carcinoma is a high incidence rate in China, which is related to high sperm preservation rate, different from other countries. The most percentage of males receiving sperm cryopreservation in the testicular cancers (15-39 years old) of China in 2020 was 5.55%, 1.29% in the lymphoma, and 0.39% in the leukemia. According to the type of cancer, a statistically significant lower pre-sperm density, total sperm output, and post-sperm density was observed in testicular cancers. It is worth noting that the prevalence of azoospermia 22.2% in leukemia patients attribute to urgent treatment before sperm cryopreservation. Disposition of cryopreserved sperm categories included continued storage (47.2%), discarded (9%), death (0.9%), and use (3.7%). CONCLUSION This study provides the first comprehensive national statistical census and review of fertility preservation in male cancer patients with respect to trends, prevalence, and cancer types. The development of male cancer fertility preservation in China is imbalanced and percentage of males receiving sperm cryopreservation in the adolescent and young adult cancers was low. Sixteen human sperm banks from China analyze current problems and challenges, and then prioritize steps toward the achievement of the FP strategy framework for Healthy China 2030.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Qiling Wang
- NHC Key Laboratory of Male Reproduction and Genetics Guangdong Provincial Reproductive, Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, 510062, Guangdong, China
| | - Huiqiang Sheng
- Zhejiang Mater Child and Reproductive Health Center, Hangzhou, 310000, Zhejiang, China
| | - Xiaowei Liang
- Human Sperm Bank of National Research Institute for Family Planning, Beijing, 100098, China
| | - Zhiqiang Wang
- The First Affiliated Hospital of Guangxi Medical University, Guilin, 530021, Guangxi, China
| | - Tianqing Meng
- Hubei Province Human Sperm Bank, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430010, Hubei, China
| | - Yushan Li
- Henan Human Sperm Bank, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, Henan, China
| | - Hao Dong
- Department of Urological Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Wenbing Zhu
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, 410008, Hunan, China
| | - Jigao Yang
- Chongqing Research Institute for Population and Family Planning Science and Technology, Human Sperm Bank, Chongqing, 401121, China
| | - Zhou Zhang
- Northwest Women's and Children's Hospital, Xian, 610045, Shaanxi, China
| | - Xianglong Jiang
- Nanchang Reproductive Hospital, Nanchang, 330001, Jiangxi, China
| | - Aiping Zhang
- Human Sperm Bank of The First Hospital of Lanzhou University, Lanzhou, 730099, Gansu, China
| | - Zuowen Liang
- The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Xiaojin He
- Anhui Provincial Human Sperm Bank, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Chunying Song
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Sperm Bank, Taiyuan, 030001, Shanxi, China
| | - Fuping Li
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.
| | - Xinzong Zhang
- NHC Key Laboratory of Male Reproduction and Genetics Guangdong Provincial Reproductive, Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, 510062, Guangdong, China.
| |
Collapse
|
5
|
Fu L, Fang F, Guo Y, Ma J, Wang S, Gu Y, Yan X, Lu W, Liu Y. Combined Analysis of the Transcriptome, Proteome and Metabolome in Human Cryopreserved Sperm. World J Mens Health 2024; 42:610-619. [PMID: 38164029 PMCID: PMC11216965 DOI: 10.5534/wjmh.230091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/15/2023] [Accepted: 07/14/2023] [Indexed: 01/03/2024] Open
Abstract
PURPOSE This study aimed to identify the altered pathways and genes associated with freezing damage in human sperm during cryopreservation by multiomics analysis. MATERIALS AND METHODS Fifteen fresh human semen samples were collected for transcriptomic analysis, and another 5 fresh human semen samples were obtained for metabolomic analysis. For each semen sample, 1 mL was cryopreserved, and another 1 mL was left untreated for paired design. The results were then combined with previously published proteomic results to identify key genes/pathways. RESULTS Cryopreservation significantly reduced sperm motility and mitochondrial structure. Transcriptomic analysis revealed altered mitochondrial function, including changes in tRNA-methyltransferase activity and adenosine tri-phosphate/adenosine di-phosphate transmembrane transporter activity. Metabolomic analysis showed that the citrate cycle in mitochondria was significantly altered. Combining transcriptomic, proteomic, and metabolomic analyses revealed 346 genes that were altered in at least two omics analyses. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that metabolic pathways were significantly altered and strongly associated with mitochondria. Five genes were altered in all three omics analyses: COL11A1, COL18A1, LPCAT3, NME1, and NNT. CONCLUSIONS Five genes were identified by multiomics analysis in human cryopreserved sperm. These genes might have specific functions in cryopreservation. Explorations of the functions of these genes will be helpful for sperm cryopreservation and sperm motility improvement or even for reproduction in the future.
Collapse
Affiliation(s)
- Longlong Fu
- National Health Commission Key Laboratory of Male Reproductive Health, Human Sperm Bank, National Research Institute for Family Planning, Beijing, China
| | - Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Ying Guo
- National Health Commission Key Laboratory of Male Reproductive Health, Human Sperm Bank, National Research Institute for Family Planning, Beijing, China
| | - Jing Ma
- Key Laboratory of Reproductive Medicine of Hebei Provincial, Hebei Research Institute of Reproductive Health, Shijiazhuang, China
| | - Shusong Wang
- Key Laboratory of Reproductive Medicine of Hebei Provincial, Hebei Research Institute of Reproductive Health, Shijiazhuang, China
| | - Yiqun Gu
- National Health Commission Key Laboratory of Male Reproductive Health, Human Sperm Bank, National Research Institute for Family Planning, Beijing, China
| | - Xiangming Yan
- Department of Pediatric Urology, Children's Hospital of Soochow University, Suzhou, China
| | - Wenhong Lu
- National Health Commission Key Laboratory of Male Reproductive Health, Human Sperm Bank, National Research Institute for Family Planning, Beijing, China.
| | - Ying Liu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
6
|
Link-Rachner CS, Göbel A, Jaschke NP, Rachner TD. Endocrine health in survivors of adult-onset cancer. Lancet Diabetes Endocrinol 2024; 12:350-364. [PMID: 38604215 DOI: 10.1016/s2213-8587(24)00088-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 04/13/2024]
Abstract
Long-term survivors of cancer (ie, the patient who is considered cured or for whom the disease is under long-term control and unlikely to recur) are at an increased risk of developing endocrine complications such as hypothalamic-pituitary dysfunctions, hypogonadisms, osteoporosis, or metabolic disorders, particularly when intensive tumour-directed therapies are applied. Symptom severity associated with these conditions ranges from mild and subclinical to highly detrimental, affecting individual health and quality of life. Although they are usually manageable, many of these endocrine pathologies remain underdiagnosed and untreated for years. To address this challenge, a higher degree of awareness, standardised screening tools, comprehensible treatment algorithms, and a close collaborative effort between endocrinologists and oncologists are essential to early identify patients who are at risk, and to implement appropriate treatment protocols. This Review highlights common symptoms and conditions related to endocrine disorders among survivors of adult-onset cancer, provides a summary of the currently available practice guidelines, and proposes a practical approach to diagnose affected patients among this group.
Collapse
Affiliation(s)
- Cornelia S Link-Rachner
- Division of Haematology and Oncology, Department of Medicine I, Technische Universität Dresden, Dresden, Germany
| | - Andy Göbel
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nikolai P Jaschke
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Tilman D Rachner
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
7
|
Tang S, Jones C, Davies J, Lane S, Mitchell RT, Coward K. Determining the optimal time interval between sample acquisition and cryopreservation when processing immature testicular tissue to preserve fertility. Cryobiology 2024; 114:104841. [PMID: 38104854 DOI: 10.1016/j.cryobiol.2023.104841] [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: 09/17/2023] [Revised: 11/11/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
The cryopreservation of immature testicular tissue (ITT) prior to gonadotoxic therapy is crucial for fertility preservation in prepubertal boys with cancer. However, the optimal holding time between tissue collection and cryopreservation has yet to be elucidated. Using the bovine model, we investigated four holding times (1, 6, 24, and 48 h) for ITTs before cryopreservation. Biopsies from two-week-old calves were stored in transport medium and cryopreserved following a standard slow-freezing clinical protocol. Thawed samples were then assessed for viability, morphology, and gene expression by haematoxylin and eosin (H&E) staining, immunohistochemistry and real-time quantitative reverse transcription-polymerase chain reaction (RT-qPCR). Analysis failed to identify any significant changes in cell viability when compared between the different groups. Sertoli (Vimentin+) and proliferating cells (Ki67+) were well-preserved. The expression of genes related to germ cells, spermatogenesis (STRA8, PLZF, GFRα-1, C-KIT, THY1, UCHL-1, NANOG, OCT-4, CREM), and apoptosis (HSP70-2) remained stable over 48 h. However, seminiferous cord detachment increased significantly in the 48-h group (p < 0.05), with associated cord and SSC shrinkage. Collectively, our analyses indicate that bovine ITTs can be stored for up to 48 h prior to cryopreservation with no impact on cell viability and the expression levels of key genes. However, to preserve the morphology of frozen-thawed tissue, the ideal processing time would be within 24 h. Testicular tissues obtained from patients for fertility preservation often need to be transported over long distances to be cryopreserved in specialist centres. Our findings highlight the importance of determining optimal tissue transport times to ensure tissue quality in cryopreservation.
Collapse
Affiliation(s)
- Shiyan Tang
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, United Kingdom; Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Celine Jones
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jill Davies
- Oxford Cell and Tissue Biobank, Children's Hospital Oxford, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Sheila Lane
- Department of Paediatric Oncology and Haematology, Children's Hospital Oxford, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom; Department of Paediatric Endocrinology, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| | - Kevin Coward
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, United Kingdom.
| |
Collapse
|
8
|
Tang S, Jones C, Mecca R, Davies J, Lane S, Coward K. An in vitrothree-dimensional (3D) testicular organoid culture system for efficient gonocyte maintenance and propagation using frozen/thawed neonatal bovine testicular tissues. Biomed Mater 2024; 19:025040. [PMID: 38324899 DOI: 10.1088/1748-605x/ad2709] [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: 09/14/2023] [Accepted: 02/07/2024] [Indexed: 02/09/2024]
Abstract
Fertility preservation in prepubertal boys with cancer requires the cryopreservation of immature testicular tissues (ITTs) prior to gonadotoxic treatment. However, the limited number of germ cells in small human ITT biopsies necessitates the development of anin vitroculture system for germ cell expansion using frozen-thawed ITTs. Here, we generated testicular organoids for thein vitromaintenance and expansion of gonocytes from frozen-thawed two-week-old neonatal bovine ITTs. We investigated the effects of different cell-seeding densities, culture serums, seeding methods, and gonadotropin supplementations, on the maintenance and proliferation of enriched gonocytes. Our results demonstrated that enriched gonocytes and testicular cells from frozen-thawed neonatal ITTs could self-assemble into spheroid organoids in three days in an appropriate Matrigel-based culture environment. For the optimal formation of prepubertal testicular organoids, a seeding density of 1 × 106cells/well is recommended over other densities. This strategy results in organoids with a mean diameter of 60.53 ± 12.12 μm; the mean number of organoids was 5.57 ± 1.60/105μm2on day 11. The viability of organoids was maintained at 79.75 ± 2.99% after being frozen and thawed. Supplementing the culture medium with glial cell-derived neurotrophic factor, fibroblast growth factor 2, and leukemia inhibitory factor, increased the proportion of KI67-positive proliferating cells in organoids, elevated the expression ofC-KITbut reduced the expression ofGFRα1at day 28 when compared to those without hormone supplements(p< 0.05). In addition, supplementing the culture medium with follicle-stimulating hormone and testosterone helped to maintain a significantly higher viability (p< 0.05) in ITT organoids at day 28. These organoids could be cryopreserved for storage and thawed as needed. The successful generation of ITT organoids provides a valuable tool for establishingin vitrospermatogenesis, propagating human germ cells, investigating testicular physiology and the origin of germ cell tumors, and testing the toxicity of new drugs in future clinical applications.
Collapse
Affiliation(s)
- Shiyan Tang
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford OX3 0AG, United Kingdom
- Present address: Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Celine Jones
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford OX3 0AG, United Kingdom
| | - Rajwa Mecca
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford OX3 0AG, United Kingdom
| | - Jill Davies
- Oxford Cell and Tissue Biobank, Children's Hospital Oxford, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, United Kingdom
| | - Sheila Lane
- Department of Paediatric Oncology and Haematology, Children's Hospital Oxford, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, United Kingdom
| | - Kevin Coward
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford OX3 0AG, United Kingdom
| |
Collapse
|
9
|
Santana-Plata A, Rivera-Egea R, Garrido N. Semen cryopreservation for an oncological reason: a retrospective study. Reprod Biomed Online 2024:103898. [PMID: 38744584 DOI: 10.1016/j.rbmo.2024.103898] [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: 07/13/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 05/16/2024]
Abstract
RESEARCH QUESTION How do cancer type and treatment affect semen quality before and after treatment, and what effect does it have in their clinical management of infertility? Also, what is the rate of patients using cryopreserved semen samples after treatment? DESIGN Patients who cryopreserved spermatozoa for oncological reasons between 2000 and 2022 in IVI clinics in Spain were retrospectively reviewed. Semen parameters were analysed before and after treatment, and utilization and destruction rates were calculated. Total motile sperm count (TMSC) was used for assisted reproductive technology (ART) counselling. RESULTS A total of 724 patients cryopreserved their semen during the study period. The semen parameters of the cancer patients' semen before and after treatment were heterogeneous, with significant differences between cancer type and semen parameters. The utilization rate was relatively low (0.4%), whereas the destruction rate was 23.1%. CONCLUSION Cancer and antineoplastic treatment affect everyone differently. Therefore, sperm cryopreservation should be offered to all patients before starting treatment to ensure their reproductive future. Furthermore, in addition to considering the semen parameters defined by the World Health Organization, it is important to use TMSC in the diagnosis of men to choose appropriate ART according to type of cancer.
Collapse
Affiliation(s)
- Ana Santana-Plata
- IVIRMA Global Research Alliance, IVI Foundation, Health Research Institute La Fe, Av. Fernando Abril Martorell, n°106, Torre A, Planta 1ª, 46026, Valencia, Spain..
| | - Rocio Rivera-Egea
- IVIRMA Global Research Alliance, Andrology Unit, IVIRMA Valencia, Plaza Policía Local, 3, 46015 Valencia, Spain
| | - Nicolás Garrido
- IVIRMA Global Research Alliance, IVI Foundation, Health Research Institute La Fe, Av. Fernando Abril Martorell, n°106, Torre A, Planta 1ª, 46026, Valencia, Spain
| |
Collapse
|
10
|
Tachibana R, Takeuchi H, Yoshikawa-Terada K, Maezawa T, Nishioka M, Takayama E, Tanaka H, Tanaka K, Hyon SH, Gen Y, Kondo E, Ikeda T. Carboxylated Poly-L-lysine Potentially Reduces Human Sperm DNA Fragmentation after Freeze-Thawing, and Its Function Is Enhanced by Low-Dose Resveratrol. Cells 2023; 12:2585. [PMID: 37998320 PMCID: PMC10670029 DOI: 10.3390/cells12222585] [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: 09/18/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023] Open
Abstract
Sperm DNA fragmentation (SDF) that occurs during the freezing-thawing of sperm may negatively impact the treatment outcomes of assisted reproductive technologies (ART). In a previous study, we developed a human sperm cryopreservation reagent containing carboxylated poly-L-lysine (CPLL) that reduced SDF after freeze-thawing compared with clinically popular cryopreservation reagents containing human serum albumin. However, it is unclear whether CPLL reduces SDF, as it differed from the constituents of the commercial cryopreservation reagents used for comparison. Therefore, here, we examined whether CPLL reduces the SDF of human sperm and evaluated reactive oxygen species (ROS) levels and lipid peroxidation (LPO), which are the causes of SDF; mitochondrial injury, ROS production; and impaired sperm motility. Furthermore, optimal antioxidants and their concentrations that could further enhance the reduction in SDF were determined for future clinical application in ART and underwent the same functional evaluations. CPLL can reduce SDF via inhibition of intracytoplasmic ROS and LPO. Furthermore, the addition of 0.1 mM resveratrol avoided the enhancement of SDF, which potentially affects mitochondrial and cytoplasmic ROS and LPO. This novel human sperm cryopreservation reagent containing CPLL and resveratrol has the potential to improve treatment outcomes in ART using frozen sperm.
Collapse
Affiliation(s)
- Ryota Tachibana
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Mie University, 2-174 Edo-bashi, Tsu 514-8507, Japan; (R.T.); (K.Y.-T.); (T.M.); (M.N.); (E.K.); (T.I.)
- Center of Advanced Reproductive Medicine, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan;
| | - Hiroki Takeuchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Mie University, 2-174 Edo-bashi, Tsu 514-8507, Japan; (R.T.); (K.Y.-T.); (T.M.); (M.N.); (E.K.); (T.I.)
- Center of Advanced Reproductive Medicine, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan;
| | - Kento Yoshikawa-Terada
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Mie University, 2-174 Edo-bashi, Tsu 514-8507, Japan; (R.T.); (K.Y.-T.); (T.M.); (M.N.); (E.K.); (T.I.)
- Center of Advanced Reproductive Medicine, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan;
| | - Tadashi Maezawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Mie University, 2-174 Edo-bashi, Tsu 514-8507, Japan; (R.T.); (K.Y.-T.); (T.M.); (M.N.); (E.K.); (T.I.)
- Center of Advanced Reproductive Medicine, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan;
| | - Mikiko Nishioka
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Mie University, 2-174 Edo-bashi, Tsu 514-8507, Japan; (R.T.); (K.Y.-T.); (T.M.); (M.N.); (E.K.); (T.I.)
- Center of Advanced Reproductive Medicine, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan;
- Obstetrics and Gynecology, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan; (H.T.); (K.T.)
| | - Erina Takayama
- Center of Advanced Reproductive Medicine, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan;
- Obstetrics and Gynecology, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan; (H.T.); (K.T.)
| | - Hiroaki Tanaka
- Obstetrics and Gynecology, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan; (H.T.); (K.T.)
| | - Kayo Tanaka
- Obstetrics and Gynecology, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan; (H.T.); (K.T.)
| | - Suong-hyu Hyon
- BMG, Inc., 45 Minamimatsunoki-cho, Higashikujo, Minami-ku, Kyoto 601-8023, Japan; (S.-h.H.); (Y.G.)
| | - Yuki Gen
- BMG, Inc., 45 Minamimatsunoki-cho, Higashikujo, Minami-ku, Kyoto 601-8023, Japan; (S.-h.H.); (Y.G.)
| | - Eiji Kondo
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Mie University, 2-174 Edo-bashi, Tsu 514-8507, Japan; (R.T.); (K.Y.-T.); (T.M.); (M.N.); (E.K.); (T.I.)
- Center of Advanced Reproductive Medicine, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan;
- Obstetrics and Gynecology, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan; (H.T.); (K.T.)
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Mie University, 2-174 Edo-bashi, Tsu 514-8507, Japan; (R.T.); (K.Y.-T.); (T.M.); (M.N.); (E.K.); (T.I.)
- Center of Advanced Reproductive Medicine, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan;
- Obstetrics and Gynecology, Mie University Hospital, 2-174 Edobashi, Tsu 514-8507, Japan; (H.T.); (K.T.)
| |
Collapse
|
11
|
Chen L, Dong Z, Chen X. Fertility preservation in pediatric healthcare: a review. Front Endocrinol (Lausanne) 2023; 14:1147898. [PMID: 37206440 PMCID: PMC10189781 DOI: 10.3389/fendo.2023.1147898] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/12/2023] [Indexed: 05/21/2023] Open
Abstract
Survival rates for children and adolescents diagnosed with malignancy have been steadily increasing due to advances in oncology treatments. These treatments can have a toxic effect on the gonads. Currently, oocyte and sperm cryopreservation are recognized as well-established and successful strategies for fertility preservation for pubertal patients, while the use of gonadotropin-releasing hormone agonists for ovarian protection is controversial. For prepubertal girls, ovarian tissue cryopreservation is the sole option. However, the endocrinological and reproductive outcomes after ovarian tissue transplantation are highly heterogeneous. On the other hand, immature testicular tissue cryopreservation remains the only alternative for prepubertal boys, yet it is still experimental. Although there are several published guidelines for navigating fertility preservation for pediatric and adolescent patients as well as transgender populations, it is still restricted in clinical practice. This review aims to discuss the indications and clinical outcomes of fertility preservation. We also discuss the probably effective and efficient workflow to facilitate fertility preservation.
Collapse
Affiliation(s)
- Lin Chen
- Reproductive Medical Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zirui Dong
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
- The Fertility Preservation Research Center, Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
12
|
Antonouli S, Di Nisio V, Messini C, Daponte A, Rajender S, Anifandis G. A comprehensive review and update on human fertility cryopreservation methods and tools. Front Vet Sci 2023; 10:1151254. [PMID: 37143497 PMCID: PMC10151698 DOI: 10.3389/fvets.2023.1151254] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/29/2023] [Indexed: 05/06/2023] Open
Abstract
The broad conceptualization of fertility preservation and restoration has become already a major concern in the modern western world since a large number of individuals often face it in the everyday life. Driven by different health conditions and/or social reasons, a variety of patients currently rely on routinely and non-routinely applied assisted reproductive technologies, and mostly on the possibility to cryopreserve gametes and/or gonadal tissues for expanding their reproductive lifespan. This review embraces the data present in human-focused literature regarding the up-to-date methodologies and tools contemporarily applied in IVF laboratories' clinical setting of the oocyte, sperm, and embryo cryopreservation and explores the latest news and issues related to the optimization of methods used in ovarian and testicular tissue cryopreservation.
Collapse
Affiliation(s)
- Sevastiani Antonouli
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Valentina Di Nisio
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Christina Messini
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| | - Alexandros Daponte
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| | - Singh Rajender
- Division of Endocrinology, Central Drug Research Institute, Lucknow, India
| | - George Anifandis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| |
Collapse
|
13
|
Kermani T, Hosseini SF, Talaei-Khozani T, Aliabadi E. Effect of Pre-Incubation of Cryopreserved Sperm with either Kisspeptin or Glutathione to Mitigate Freeze-Thaw Damage. IRANIAN JOURNAL OF MEDICAL SCIENCES 2023; 48:198-208. [PMID: 36895454 PMCID: PMC9989238 DOI: 10.30476/ijms.2022.92300.2354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/11/2022] [Accepted: 01/31/2022] [Indexed: 03/11/2023]
Abstract
Background Sperm cryopreservation reduces sperm quality. Kisspeptin (KP) has beneficial effects on sperm functions. This study compares the effect of KP and Glutathione (GSH) on mitigating the detrimental effects of the freeze-thaw cycle on sperm. Methods An experimental study was conducted in Birjand (Iran) during 2018-2020. Thirty normal swim-up semen samples were treated with Ham's F10 medium (negative control), 1 mM GSH (positive control), or KP (10 µM) for 30 min before freezing. The motility, acrosome reaction, capacitation, and DNA quality of the frozen-thawed sperms were assessed according to the WHO guidelines. Statistical analysis was performed using paired t test, one-way analysis of variance, and least significant difference. Results Pre-incubation with KP significantly increased the percentage of sperm motility (34.00±6.7, P=0.003) compared to the control (20.44±7.4) and GSH-treated (31.25±12.2) aliquots. The frequency of non-capacitated spermatozoa was significantly higher in the KP-treated group (98.73%) than in the control (96.46%) and GSH-treated (96.49%) aliquots (P<0.001). The percentage of acrosome-intact spermatozoa in the KP-treated group (77.44%) was significantly higher than the control (74.3%) and GSH-treated (74.54%) groups (P<0.001). The sperm frequency with normal histone in the KP-treated group (51.86%) and with normal protamine (65.39%) was significantly higher than the controls (P=0.001 and P=0.002, respectively). The percentage of TUNEL-positive sperm was significantly lower in the KP-treated group (9.09±2.71) than both GSH-treated (11.22±2.73) and control (11.31±2.2) groups (both P=0.002). Conclusion Pre-incubation with KP protects sperm motility and DNA integrity from the detrimental effect of the freeze-thaw cycle. KP is suitable as a pre-treatment to control sperm quality during freezing-thawing.
Collapse
Affiliation(s)
- Tayebeh Kermani
- Department of Anatomy, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Syedeh-Fatemeh Hosseini
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Talaei-Khozani
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Aliabadi
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
14
|
Agarwal MS, Kelly KB, Papanikolaou P, Kirby N, Ha CS. Unique approaches for testicular shielding during total-body irradiation for pediatric patients. J Appl Clin Med Phys 2022; 24:e13842. [PMID: 36355034 PMCID: PMC9859990 DOI: 10.1002/acm2.13842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 11/12/2022] Open
Abstract
Total-body irradiation (TBI) has been used as a part of the conditioning regimen for patients undergoing hematopoietic stem cell transplantation for certain nonmalignant conditions such as sickle cell disease. Although effective, TBI can cause lasting side effects for pediatric patients. One of these potential side effects includes oligospermia or even permanent azoospermia. Although many investigators have studied ways to shield the testicles during the TBI for nonmalignant conditions, there is no set standard. We describe the technical aspects of effective techniques to shield the testicles of male pediatric patients undergoing TBI. We verified that our techniques reduced the testicular dose by approximately 80%-85% of the TBI prescription dose in four male pediatric patients, keeping the dose well below the documented doses that can cause permanent infertility and hypogonadism.
Collapse
Affiliation(s)
- Mohit Shiv Agarwal
- Department of Radiation Oncology, Mays Cancer CenterUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Kathryn Bales Kelly
- Department of Radiation Oncology, Mays Cancer CenterUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Panos Papanikolaou
- Department of Radiation Oncology, Mays Cancer CenterUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Neil Kirby
- Department of Radiation Oncology, Mays Cancer CenterUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Chul Soo Ha
- Department of Radiation Oncology, Mays Cancer CenterUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| |
Collapse
|
15
|
Whelan EC, Yang F, Avarbock MR, Sullivan MC, Beiting DP, Brinster RL. Reestablishment of spermatogenesis after more than 20 years of cryopreservation of rat spermatogonial stem cells reveals an important impact in differentiation capacity. PLoS Biol 2022; 20:e3001618. [PMID: 35536782 PMCID: PMC9089916 DOI: 10.1371/journal.pbio.3001618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 04/04/2022] [Indexed: 12/22/2022] Open
Abstract
Treatment of cancer in children is increasingly successful but leaves many prepubertal boys suffering from infertility or subfertility later in life. A current strategy to preserve fertility in these boys is to cryopreserve a testicular biopsy prior to treatment with the expectation of future technologies allowing for the reintroduction of stem cells and restoration of spermatogenesis. Spermatogonial stem cells (SSCs) form the basis of male reproduction, differentiating into all germ cell types, including mature spermatozoa and can regenerate spermatogenesis following transplantation into an infertile testis. Here, we demonstrate that rat SSCs frozen for more than 20 years can be transplanted into recipient mice and produce all differentiating germ cell types. However, compared with freshly isolated cells or those frozen for a short period of time, long-frozen cells do not colonize efficiently and showed reduced production of spermatids. Single-cell RNA sequencing revealed similar profiles of gene expression changes between short- and long-frozen cells as compared with fresh immediately after thawing. Conversely, following transplantation, long-frozen samples showed enhanced stem cell signaling in the undifferentiated spermatogonia compartment, consistent with self-renewal and a lack of differentiation. In addition, long-frozen samples showed fewer round spermatids with detectable protamine expression, suggesting a partial block of spermatogenesis after meiosis resulting in a lack of elongating spermatids. These findings strongly suggest that prolonged cryopreservation can impact the success of transplantation to produce spermatogenesis, which may not be revealed by analysis of the cells immediately after thawing. Our analysis uncovered persistent effects of long-term freezing not found in other cryopreservation studies that lacked functional regeneration of the tissue and this phenomenon must be accounted for any future therapeutic application. This study shows that spermatogonial stem cells frozen for more than 20 years can repopulate the niche when transplanted into an infertile host, but with lower efficiency and displaying abnormal spermatogenesis compared to fresh or short-term frozen cells. Single-cell RNA sequencing reveals details of the dysregulated signaling.
Collapse
Affiliation(s)
- Eoin C. Whelan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Fan Yang
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Histology and Embryology, Medical College, Yangzhou University, Yangzhou, China
| | - Mary R. Avarbock
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Megan C. Sullivan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ralph L. Brinster
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
16
|
Abdel-Latif R, Fathy M, Anwar HA, Naseem M, Dandekar T, Othman EM. Cisplatin-Induced Reproductive Toxicity and Oxidative Stress: Ameliorative Effect of Kinetin. Antioxidants (Basel) 2022; 11:antiox11050863. [PMID: 35624727 PMCID: PMC9137797 DOI: 10.3390/antiox11050863] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 12/13/2022] Open
Abstract
Cisplatin is a commonly used chemotherapeutic agent; however, its potential side effects, including gonadotoxicity and infertility, are a critical problem. Oxidative stress has been implicated in the pathogenesis of cisplatin-induced testicular dysfunction. We investigated whether kinetin use at different concentrations could alleviate gonadal injury associated with cisplatin treatment, with an exploration of the involvement of its antioxidant capacity. Kinetin was administered in different doses of 0.25, 0.5, and 1 mg/kg, alone or along with cisplatin for 10 days. Cisplatin toxicity was induced via a single IP dose of 7 mg/kg on day four. In a dose-dependent manner, concomitant administration of kinetin with cisplatin significantly restored testicular oxidative stress parameters, corrected the distorted sperm quality parameters and histopathological changes, enhanced levels of serum testosterone and testicular StAR protein expression, as well as reduced the up-regulation of testicular TNF-α, IL-1β, Il-6, and caspase-3, caused by cisplatin. It is worth noting that the testicular protective effect of the highest kinetin dose was comparable/more potent and significantly higher than the effects of vitamin C and the lowest kinetin dose, respectively. Overall, these data indicate that kinetin may offer a promising approach for alleviating cisplatin-induced reproductive toxicity and organ damage, via ameliorating oxidative stress and reducing inflammation and apoptosis.
Collapse
Affiliation(s)
- Rania Abdel-Latif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Minia, Minia 61519, Egypt;
| | - Moustafa Fathy
- Department of Biochemistry, Faculty of Pharmacy, University of Minia, Minia 61519, Egypt; (M.F.); (H.A.A.)
| | - Hend Ali Anwar
- Department of Biochemistry, Faculty of Pharmacy, University of Minia, Minia 61519, Egypt; (M.F.); (H.A.A.)
| | - Muhammad Naseem
- Department of Life and Environmental Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi 144534, United Arab Emirates;
- Department of Bioinformatics, Biocenter, Am Hubland University of Wuerzburg, 97074 Wuerzburg, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, Am Hubland University of Wuerzburg, 97074 Wuerzburg, Germany
- Correspondence: (T.D.); (E.M.O.)
| | - Eman M. Othman
- Department of Biochemistry, Faculty of Pharmacy, University of Minia, Minia 61519, Egypt; (M.F.); (H.A.A.)
- Department of Bioinformatics, Biocenter, Am Hubland University of Wuerzburg, 97074 Wuerzburg, Germany
- Correspondence: (T.D.); (E.M.O.)
| |
Collapse
|
17
|
Kabiri D, Safrai M, Gropp M, Hidas G, Mordechai-Daniel T, Meir K, Revel A, Imbar T, Reubinoff B. Establishment of a controlled slow freezing-based approach for experimental clinical cryopreservation of human prepubertal testicular tissues. F S Rep 2022; 3:47-56. [PMID: 35386499 PMCID: PMC8978083 DOI: 10.1016/j.xfre.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- Doron Kabiri
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Myriam Safrai
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Michal Gropp
- The Sydney and Judy Swartz Embryonic Stem Cell Research Center of The Goldyne Savad Institution of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Guy Hidas
- Department of Pediatric Urology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | - Karen Meir
- Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ariel Revel
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Tal Imbar
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- IVF unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Benjamin Reubinoff
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- The Sydney and Judy Swartz Embryonic Stem Cell Research Center of The Goldyne Savad Institution of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- IVF unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- Reprint requests: Benjamin Reubinoff, M.D., Ph.D., Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
18
|
Corda PO, Silva JV, Pereira SC, Barros A, Alves MG, Fardilha M. Bioinformatic Approach to Unveil Key Differentially Expressed Proteins in Human Sperm After Slow and Rapid Cryopreservation. Front Cell Dev Biol 2022; 9:759354. [PMID: 35145967 PMCID: PMC8821918 DOI: 10.3389/fcell.2021.759354] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/23/2021] [Indexed: 11/25/2022] Open
Abstract
Currently, two conventional freezing techniques are used in sperm cryopreservation: slow freezing (SF) and rapid freezing (RF). Despite the protocolar improvements, cryopreservation still induces significant alterations in spermatozoon that are poorly understood. Here, available proteomic data from human cryopreserved sperm was analyzed through bioinformatic tools to unveil key differentially expressed proteins (DEPs) that can be used as modulation targets or quality markers. From the included proteomic studies, 160 and 555 DEPs were collected for SF and RF groups, respectively. For each group, an integrative network was constructed using gene ontology and protein-protein interaction data to identify key DEPs. Among them, arylsulfatase A (ARSA) was highlighted in both freezing networks, and low ARSA levels have been associated with poor-sperm quality. Thus, ARSA was selected for further experimental investigation and its levels were assessed in cryopreserved samples by western blot. ARSA levels were significantly decreased in RF and SF samples (∼31.97 and ∼39.28%, respectively). The bioinformatic analysis also revealed that the DEPs were strongly associated with proteasomal and translation pathways. The purposed bioinformatic approach allowed the identification of potential key DEPs in freeze-thawed human spermatozoa. ARSA has the potential to be used as a marker to assess sperm quality after cryopreservation.
Collapse
Affiliation(s)
- Pedro O Corda
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Joana Vieira Silva
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal.,Department of Chemistry, QOPNA and LAQV, University of Aveiro, Aveiro, Portugal.,Clinical and Experimental Endocrinology, Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Sara C Pereira
- Clinical and Experimental Endocrinology, Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Alberto Barros
- Department of Chemistry, QOPNA and LAQV, University of Aveiro, Aveiro, Portugal.,Centre for Reproductive Genetics A. Barros, Porto, Portugal.,Department of Genetics, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Marco G Alves
- Clinical and Experimental Endocrinology, Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Margarida Fardilha
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| |
Collapse
|
19
|
Abstract
Patient survival following childhood cancer has increased with contemporary radiation and chemotherapy techniques. However, gonadotoxicity associated with treatments means that infertility is a common consequence in survivors. Novel fertility preservation options are emerging, but knowledge about these options amongst urologists and other medical professionals is lacking. Pre-pubertal boys generally do not produce haploid germ cells. Thus, strategies for fertility preservation require cryopreservation of tissue containing spermatogonial stem cells (SSCs). Few centres worldwide routinely offer this option and fertility restoration (including testicular tissue engraftment, autotransplantation of SSCs and in vitro maturation of SSCs to spermatozoa) post-thaw is experimental. In pubertal boys, the main option for fertility preservation is masturbation and cryopreservation of the ejaculate. Assisted ejaculation using penile vibratory stimulation or electroejaculation and surgical sperm retrieval can be used in a sequential manner after failed masturbation. Physicians should inform boys and parents about the gonadotoxic effects of cancer treatment and offer fertility preservation. Preclinical experience has identified challenges in pre-pubertal fertility preservation, but available options are expected to be successful when today's pre-pubertal boys with cancer become adults. By contrast, fertility preservation in pubertal boys is clinically proven and should be offered to all patients undergoing cancer treatment.
Collapse
|
20
|
Harada M, Kimura F, Takai Y, Nakajima T, Ushijima K, Kobayashi H, Satoh T, Tozawa A, Sugimoto K, Saji S, Shimizu C, Akiyama K, Bando H, Kuwahara A, Furui T, Okada H, Kawai K, Shinohara N, Nagao K, Kitajima M, Suenobu S, Soejima T, Miyachi M, Miyoshi Y, Yoneda A, Horie A, Ishida Y, Usui N, Kanda Y, Fujii N, Endo M, Nakayama R, Hoshi M, Yonemoto T, Kiyotani C, Okita N, Baba E, Muto M, Kikuchi I, Morishige KI, Tsugawa K, Nishiyama H, Hosoi H, Tanimoto M, Kawai A, Sugiyama K, Boku N, Yonemura M, Hayashi N, Aoki D, Osuga Y, Suzuki N. Japan Society of Clinical Oncology Clinical Practice Guidelines 2017 for fertility preservation in childhood, adolescent, and young adult cancer patients: part 1. Int J Clin Oncol 2022; 27:265-280. [PMID: 34973107 PMCID: PMC8816532 DOI: 10.1007/s10147-021-02081-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/07/2021] [Indexed: 11/30/2022]
Abstract
In 2017, the Japan Society of Clinical Oncology (JSCO) published the JSCO Clinical Practice Guidelines 2017 for Fertility Preservation in Childhood, Adolescent, and Young Adult Cancer Patients. These were the first Japanese guidelines to address issues of oncofertility. In this field of medicine, sustained close cooperation between oncologists and reproductive specialists is essential from the diagnosis of cancer until many years after completion of cancer treatment. These JSCO guidelines were intended to guide multidisciplinary medical staff in considering the availability of fertility preservation options and to help them decide whether to provide fertility preservation to childhood, adolescent, and young adult cancer patients before treatment starts, with the ultimate goal of improving patient survivorship. The guidelines are presented as Parts 1 and 2. This article (Part 1) summarizes the goals of the guidelines and the methods used to develop them and provides an overview of fertility preservation across all oncology areas. It includes general remarks on the basic concepts surrounding fertility preservation and explanations of the impacts of cancer treatment on gonadal function by sex and treatment modality and of the options for protecting/preserving gonadal function and makes recommendations based on 4 clinical questions. Part 2 of these guidelines provides specific recommendations on fertility preservation in 8 types of cancer (gynecologic, breast, urologic, pediatric, hematologic, bone and soft tissue, brain, and digestive).
Collapse
Affiliation(s)
- Miyuki Harada
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, 113-8655, Japan
| | - Fuminori Kimura
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Seta Tsukinowa-Cho Otsu, Shiga, 520-2192, Japan
| | - Yasushi Takai
- Department of Obstetrics and Gynecology Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan
| | - Takeshi Nakajima
- Department of Endoscopy, Gastrointestinal Endoscopy Division, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kimio Ushijima
- Department of Obstetrics and Gynecology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Hiroaki Kobayashi
- Department of Obstetrics and Gynecology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Toyomi Satoh
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tsukuba, Tennoudai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Akiko Tozawa
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| | - Kohei Sugimoto
- International Center for Reproductive Medicine, Dokkyo Medical University Saitama Medical Center, 2-1-50 Minamikoshigaya, Koshigaya, Saitama, 343-8555, Japan
| | - Shigehira Saji
- Department of Medical Oncology, Fukushima Medical University, 1 Hikarigaoka, Fukushima-city, Fukushima, 960-1295, Japan
| | - Chikako Shimizu
- Department of Breast and Medical Oncology, Comprehensive Cancer Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Kyoko Akiyama
- Department of Breast and Endocrine Surgery, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| | - Hiroko Bando
- Department of Breast and Endocrine Surgery, Faculty of Medicine, University of Tsukuba, Tennoudai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Akira Kuwahara
- Ladies Clinic Cosmos Kochi, 6-27, Sugiiru, Kochi, Kochi, 780-0082, Japan
| | - Tatsuro Furui
- Department of Obstetrics and Gynecology, Gifu University Graduate School of Medicine, 1-1, Yanagido, Gifu City, Gifu, 501-1194, Japan
| | - Hiroshi Okada
- International Center for Reproductive Medicine, Dokkyo Medical University Saitama Medical Center, 2-1-50 Minamikoshigaya, Koshigaya, Saitama, 343-8555, Japan
| | - Koji Kawai
- Department of Urology, International University of Health and Welfare, 852, Hatakeda Narita, Chiba, 286-0124, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Kita 15Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Koichi Nagao
- Department of Urology, Toho University Faculty of Medicine, 6-11-1, Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Michio Kitajima
- Department of Obstetrics and Gynecology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Souichi Suenobu
- Division of General Pediatrics and Emergency Medicine, Department of Pediatrics, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama, Yufu, Oita, 879-5593, Japan
| | - Toshinori Soejima
- Department of Radiation Oncology, Kobe Proton Center, 1-6-8, Minatojima-minamimachi, Chuo-ku, Kobe City, Hyogo, 650-0047, Japan
| | - Mitsuru Miyachi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yoko Miyoshi
- Department of Health and Nutrition, Faculty of Health and Nutrition, Osaka Shoin Women's University, 4-2-26 Hishiya-nishi, Higashi-Osaka, Osaka, 577-8550, Japan
| | - Akihiro Yoneda
- Division of Pediatric Surgical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Akihito Horie
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyoku, Kyoto, 606-8507, Japan
| | - Yasushi Ishida
- Pediatric Medical Center, Ehime Prefectural Central Hospital, 83 Kasuga-machi, Matsuyama-city, Ehime, 790-0024, Japan
| | - Noriko Usui
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Department of Medicine, Jichi Medical University, 1-847 Amanuma, Omiya-ku, Saitama-city, Saitama, 330-8503, Japan
| | - Nobuharu Fujii
- Division of Transfusion, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Makoto Endo
- Department of Orthopaedic Surgery, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Robert Nakayama
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Manabu Hoshi
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585, Japan
| | - Tsukasa Yonemoto
- Division of Orthopedic Surgery, Chiba Cancer Center, 666-2 Nitona-cho, Chuo-ku, Chiba, 260-8717, Japan
| | - Chikako Kiyotani
- Children's Cancer Center, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Natsuko Okita
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Eishi Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyoku, Kyoto, 606-8507, Japan
| | - Iwaho Kikuchi
- Department of Obstetrics and Gynecology, Medical Park Yokohama, 1-1-8, Sakuragi-cho, Yokohama, Kanagawa, 231-0062, Japan
| | - Ken-Ichirou Morishige
- Department of Obstetrics and Gynecology, Gifu University Graduate School of Medicine, 1-1, Yanagido, Gifu City, Gifu, 501-1194, Japan
| | - Koichiro Tsugawa
- Department of Breast and Endocrine Surgery, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| | - Hiroyuki Nishiyama
- Department of Urology, Faculty of Medicine, University of Tsukuba, Tennoudai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hajime Hosoi
- Department of Nursing, Doshisha Women's College of Liberal Arts, Kodo, Kyotanabe City, Kyoto, 610-0395, Japan
| | - Mitsune Tanimoto
- Chugoku Central Hospital, 148-13, Kamiiwanari, Miyuki-cho, Fukuyama-city, Hiroshima, 720-0001, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology and Rehabilitation Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kazuhiko Sugiyama
- Department of Clinical Oncology and Neuro-Oncology Program, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Narikazu Boku
- Department of Medical Oncology and General Medicine, Institute of Medical Science, IMSUT Hospital, University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Masato Yonemura
- Department of Pharmacy, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa-shi, Chiba, 277-0882, Japan
| | - Naoko Hayashi
- Graduate School of Nursing Science, St Luke's International University, 10-1 Akashi-cho, Chuo-ku, Tokyo, 104-0044, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, 113-8655, Japan.
| | - Nao Suzuki
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| |
Collapse
|
21
|
Lue Y, Swerdloff R, Jia Y, Wang C. The emerging role of mitochondrial derived peptide humanin in the testis. Biochim Biophys Acta Gen Subj 2021; 1865:130009. [PMID: 34534645 DOI: 10.1016/j.bbagen.2021.130009] [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: 06/30/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 11/19/2022]
Abstract
The discovery of mitochondrial derive peptides (MDPs) has spotlighted mitochondria as central hubs in control and regulation of cell viability and metabolism in the testis in response to intracellular and extracellular stresses. MDPs (Humanin, MOTS-c and SHLP-2) are present in testes. Humanin, the first MDP, is predominantly expressed in Leydig cells, and moderately in germ cells and seminal plasma. The administration of synthetic humanin peptide agonist HNG protects male germ cells against apoptosis induced by intratesticular hormonal deprivation, testicular hyperthermia, and chemotherapeutic agents in rodent testes. Humanin interacting with IGFBP-3 and/or Bax (pro-apoptotic proteins) prevents the activation of germ cell apoptosis. Humanin participates in the network of IL-12/IL-27 family of cytokines to exert the immune-modulation of the testicular environment. Humanin and other MDPs may be important in the amelioration of testicular stress and prevention of cell injury with possible implications for male infertility, fertility preservation and contraceptive development.
Collapse
Affiliation(s)
- Yanhe Lue
- Division of Endocrinology, Department of Medicine, The Lundquist Institute and Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Ronald Swerdloff
- Division of Endocrinology, Department of Medicine, The Lundquist Institute and Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Yue Jia
- Department of Pathology, The Lundquist Institute and Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Christina Wang
- Division of Endocrinology, Department of Medicine, The Lundquist Institute and Harbor-UCLA Medical Center, Torrance, CA, United States of America.
| |
Collapse
|
22
|
Lackamp N, Wilkemeyer I, Jelas I, Keller U, Bullinger L, Stintzing S, le Coutre P. Survey of Long-Term Experiences of Sperm Cryopreservation in Oncological and Non-Oncological Patients: Usage and Reproductive Outcomes of a Large Monocentric Cohort. Front Oncol 2021; 11:772809. [PMID: 34804976 PMCID: PMC8602360 DOI: 10.3389/fonc.2021.772809] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/19/2021] [Indexed: 11/29/2022] Open
Abstract
Progress in oncological treatment has led to an improved long-term survival of young male cancer patients over the last decades. However, standard cancer treatments frequently implicate fertility-damaging potential. Cryopreservation of sperm is the current standard option to preserve patient’s fertility after treatment, yet long-term data on usage and reproductive experiences is still limited. Natural fertility after treatment and especially in relation to the type of treatment has been poorly analyzed so far. Therefore, we performed a retrospective survey including male patients with an indication for gonadotoxic treatment who cryopreserved reproductive material at our institution between 1994 and 2017. Study questionnaires regarding treatment, material usage, and reproductive outcomes were sent to eligible patients. Additionally, semen analyses of study participants from the time of cryopreservation were evaluated. A total of 99 patients were included in the study. Respondents’ median age was 38.0 years. Most frequent diagnoses were testicular cancer (29.3%) and lymphoma (26.3%). A further 8.1% suffered from autoimmune diseases. Testicular cancer patients had a significantly lower pre-treatment median sperm concentration (18.0 million/ml) compared to non-testicular cancer patients (54.2 million/ml). Until November 2020, the determined sperm usage and cumulative live-birth rate per couple were 17.2% and 58.8%, respectively. Most sperm users received treatments with high (40.0%) or intermediate (33.3%) gonadotoxic potential. 20.7% of all patients reported to had fathered at least one naturally conceived child after treatment, this being the case especially if they had been treated with less or potentially gonadotoxic therapies. In conclusion, our findings emphasize the importance of sperm cryopreservation in the context of male fertility preservation. Furthermore, they indicate that the gonadotoxic potential of patients’ treatments could represent a predictive factor for sperm usage.
Collapse
Affiliation(s)
- Nadine Lackamp
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ina Wilkemeyer
- Department of Urology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ivan Jelas
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sebastian Stintzing
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp le Coutre
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
23
|
Astaxanthin Relieves Busulfan-Induced Oxidative Apoptosis in Cultured Human Spermatogonial Stem Cells by Activating the Nrf-2/HO-1 pathway. Reprod Sci 2021; 29:374-394. [PMID: 34129218 DOI: 10.1007/s43032-021-00651-x] [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: 03/02/2021] [Accepted: 05/30/2021] [Indexed: 12/30/2022]
Abstract
Many child cancer patients endure anticancer therapy containing alkylating agents before sexual maturity. Busulfan (BU), as an alkylating agent, is a chemotherapy drug, causing DNA damage and cytotoxicity in germ cells. In the present study, we aimed to investigate the protective effect of astaxanthin (AST), as a potent antioxidant and powerful reactive oxygen species (ROS) scavenger, on BU-induced toxicity in human spermatogonial stem cells. For this purpose, testes were obtained from four brain-dead donors. After tissue enzymatic digestions, testicular cells were cultured for 3 weeks for spermatogonial stem cell (SSC) isolation and purification. K562 cell line was cultured to survey the effect of AST on cancer treatment. The cultured SSCs and K562 cell line were finally treated with AST (10μM), BU (0.1nM), and AST+BU. The expression of NRF-2, HO-1, SOD2, SOD3, TP53, and apoptotic genes, including CASP9, CASP3, BCL2, and BAX, were assayed using real-time PCR. Moreover, ROS level in different groups and malondialdehyde level and total antioxidant capacity in cell contraction of SSCs were measured using ELISA. Data showed that AST significantly upregulated the expression of NRF-2 gene (P<0.001) and protein (P<0.005) and also significantly decreased the production of BU-induced ROS (P<0.001). AST activated the NRF-2/HO-1 pathway that could remarkably restrain BU-induced apoptosis in SSCs. Interestingly, AST upregulated the expression level of apoptosis genes in the K562 cell line. The results of this study indicated that AST reduces the side effects of BU on SSCs without interference with its chemotherapy effect on cancerous cells through modulation of the NRF-2/HO-1 and mitochondria-mediated apoptosis pathways.
Collapse
|
24
|
Pediatric and Adolescent Oncofertility in Male Patients-From Alpha to Omega. Genes (Basel) 2021; 12:genes12050701. [PMID: 34066795 PMCID: PMC8150386 DOI: 10.3390/genes12050701] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 01/15/2023] Open
Abstract
This article reviews the latest information about preserving reproductive potential that can offer enhanced prospects for future conception in the pediatric male population with cancer, whose fertility is threatened because of the gonadotoxic effects of chemotherapy and radiation. An estimated 400,000 children and adolescents aged 0–19 years will be diagnosed with cancer each year. Fertility is compromised in one-third of adult male survivors of childhood cancer. We present the latest approaches and techniques for fertility preservation, starting with fertility preservation counselling, a clinical practice guideline used around the world and finishing with recent advances in basic science and translational research. Improving strategies for the maturation of germ cells in vitro combined with new molecular techniques for gene editing could be the next scientific keystone to eradicate genetic diseases such as cancer related mutations in the offspring of cancer survivors.
Collapse
|
25
|
Reschini M, Somigliana E, Meazza C, Podda M, Guarneri C, Giacchetta D, Massimino M, Restelli L, Filippi F, Terenziani M. Sperm cryopreservation in adolescents with cancer. Eur J Obstet Gynecol Reprod Biol 2021; 260:198-202. [PMID: 33839645 DOI: 10.1016/j.ejogrb.2021.03.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate whether sperm banking is under-used in adolescents with cancer. STUDY DESIGN We reviewed our 33 years long experience of semen cryopreservation and identified male subjects with cancer aged 14-21 years who banked their sperm. We hypothesized as a referring model a constant rate of referral in every age category (uniform distribution). In addition, we evaluated whether the distribution of the referrals per age varied according to historical periods and compared semen quality in the different age groups. RESULTS Overall, 317 subjects were selected. A sharp increase with age emerged (p < 0.001): the number of cases augmented from 4 at age 14 years to 79 at age 21 years. This significant increase persisted even when focussing only on subjects older than 16-18 years. An improvement of the distribution of cases per age group emerged when limiting the analysis to the last decade of activity; however, the increase remained significant (p < 0.001). Finally, we investigated whether semen quality differed according to age groups and failed to show marked differences, suggesting that sperm banking even at the youngest ages could be of potential benefit. CONCLUSIONS This study confirmed that sperm banking is under-used in adolescents with cancers. Future studies should better clarify the determinants of this low referral and identify interventions that can improve the situation.
Collapse
Affiliation(s)
- Marco Reschini
- Infertility Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Edgardo Somigliana
- Infertility Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Dept Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Cristina Meazza
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Marta Podda
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Cristina Guarneri
- Infertility Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Giacchetta
- Assisted Reproduction Techniques Unit, Ospedale Santi Paolo e Carlo, Milan, Italy
| | - Maura Massimino
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Liliana Restelli
- Infertility Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Filippi
- Infertility Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Monica Terenziani
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
26
|
Papler TB, Vrtacnik-Bokal E, Drobnic S, Stimpfel M. The outcome of IVF/ICSI cycles in male cancer patients: retrospective analysis of procedures from 2004 to 2018. Radiol Oncol 2021; 55:221-228. [PMID: 33675201 PMCID: PMC8042825 DOI: 10.2478/raon-2021-0011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/25/2021] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION Fertility preservation is an important aspect of quality of life in oncological patients, and in men is achieved by semen cryopreservation prior to treatment. Results of in vitro fertilization (IVF) procedures in healthy infertile couples are comparable, regardless of whether fresh or cryopreserved semen is used, but are scarce in male oncological patients. PATIENTS AND METHODS We performed a retrospective analysis of IVF/intracytoplasmic sperm injection (IVF/ICSI) procedures in infertile couples where men had been treated for cancer in the past. We additionally compared the results of IVF/ICSI procedures with respect to the type of semen used (fresh, cryopreserved). RESULTS We compared the success rates of 214 IVF/ICSI cycles performed in the years 2004-2018. Pregnancy (30.0% vs. 21.4%; p = 0.12) and live-birth rates (22.3% vs. 17.9%; p = 0.43) per oocyte aspiration were similar between the groups in fresh cycles; however embryo utilization (48.9% vs. 40.0%; p = 0.006) and embryo cryopreservation rates (17.3% vs. 12.7%; p = 0.048) were significantly higher in the cryopreserved semen group. The cumulative pregnancy rate (60.6% vs. 37.7%; p = 0.012) was significantly higher, and the live-birth rate (45.1% vs. 34.0%; p = 0.21) non-significantly higher, in the cryopreserved semen group. CONCLUSIONS The success of IVF/ICSI procedures in couples where the male partner was treated for cancer in the past are the same in terms of pregnancies and live-births in fresh cycles regardless of the type of semen used. However, embryo utilization and embryo cryopreservation rates are significantly higher when cryopreserved semen is used, leading to a significantly higher cumulative number of couples who achieved at least one pregnancy.
Collapse
Affiliation(s)
- Tanja Burnik Papler
- Department of Human Reproduction, Division of Gynaecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Eda Vrtacnik-Bokal
- Department of Human Reproduction, Division of Gynaecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Saso Drobnic
- Department of Human Reproduction, Division of Gynaecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Martin Stimpfel
- Department of Human Reproduction, Division of Gynaecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
27
|
Cancer survivorship: Reproductive health outcomes should be included in standard toxicity assessments. Eur J Cancer 2021; 144:310-316. [DOI: 10.1016/j.ejca.2020.11.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/17/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022]
|
28
|
Zavattaro M, Felicetti F, Faraci D, Scaldaferri M, Dellacasa C, Busca A, Dionisi-Vici M, Cattel F, Motta G, Giaccone L, Ghigo E, Arvat E, Lanfranco F, Bruno B, Brignardello E. Impact of Allogeneic Stem Cell Transplantation on Testicular and Sexual Function. Transplant Cell Ther 2020; 27:182.e1-182.e8. [PMID: 33830036 DOI: 10.1016/j.jtct.2020.10.020] [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: 08/24/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 10/22/2022]
Abstract
High-dose chemotherapy and radiotherapy, administered as a conditioning regimen before stem cell transplantation, are known to negatively impact testicular function and sexuality. However, to date, only a few studies have simultaneously analyzed the real prevalence of these complications in this clinical setting. Therefore, this study aimed to assess the prevalence of testicular dysfunction and sexual impairment in a cohort of males who underwent allogeneic stem cell transplantation in adulthood. This observational, cross-sectional, single-center study consecutively enrolled 105 subjects on outpatient follow-up. Testicular function and sexuality were evaluated through a hormonal profile (testosterone, follicle-stimulating hormone, luteinizing hormone, and inhibin B) and the IIEF-15 questionnaire, respectively. We found a higher prevalence of hypogonadism (21%), impaired spermatogenesis (87%), and erectile dysfunction (72%) compared with the general population. Chronic graft-versus-host disease, especially of moderate/severe grade, was associated with an increased risk of developing erectile dysfunction (odds ratio, 6.338). Moreover, a high proportion of patients presented with alterations in all domains of sexual function, even after complete clinical remission of hematologic disease. Our data confirm both testicular function and sexuality alterations as frequent complications after allogeneic stem cell transplantation. A multidisciplinary approach is advisable for early diagnosis and adequate treatment.
Collapse
Affiliation(s)
- Marco Zavattaro
- Andrology Unit, Division of Endocrinology, Diabetology, and Metabolism, Città della Salute e della Scienza Hospital, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - Francesco Felicetti
- Transition Unit for Childhood Cancer Survivors, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Danilo Faraci
- Department of Oncology, Stem Cell Transplant Center, Città della Salute e della Scienza Hospital, Turin, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | | - Chiara Dellacasa
- Department of Oncology, Stem Cell Transplant Center, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Alessandro Busca
- Department of Oncology, Stem Cell Transplant Center, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Margherita Dionisi-Vici
- Transition Unit for Childhood Cancer Survivors, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Francesco Cattel
- Pharmacy, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Giovanna Motta
- Andrology Unit, Division of Endocrinology, Diabetology, and Metabolism, Città della Salute e della Scienza Hospital, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - Luisa Giaccone
- Department of Oncology, Stem Cell Transplant Center, Città della Salute e della Scienza Hospital, Turin, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Ezio Ghigo
- Andrology Unit, Division of Endocrinology, Diabetology, and Metabolism, Città della Salute e della Scienza Hospital, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - Emanuela Arvat
- Department of Medical Sciences, University of Turin, Turin, Italy; Division of Oncological Endocrinology, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Fabio Lanfranco
- Andrology Unit, Division of Endocrinology, Diabetology, and Metabolism, Città della Salute e della Scienza Hospital, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - Benedetto Bruno
- Department of Oncology, Stem Cell Transplant Center, Città della Salute e della Scienza Hospital, Turin, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Enrico Brignardello
- Transition Unit for Childhood Cancer Survivors, Città della Salute e della Scienza Hospital, Turin, Italy.
| |
Collapse
|
29
|
Gul M, Hildorf S, Dong L, Thorup J, Hoffmann ER, Jensen CFS, Sønksen J, Cortes D, Fedder J, Andersen CY, Goossens E. Review of injection techniques for spermatogonial stem cell transplantation. Hum Reprod Update 2020; 26:368-391. [PMID: 32163572 DOI: 10.1093/humupd/dmaa003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/07/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Although the prognosis of childhood cancer survivors has increased dramatically during recent years, chemotherapy and radiation treatments for cancer and other conditions may lead to permanent infertility in prepubertal boys. Recent developments have shown that spermatogonial stem cell (SSC) transplantation may be a hope for restoring fertility in adult survivors of childhood cancers. For this reason, several centres around the world are collecting and cryopreserving testicular tissue or cells anticipating that, in the near future, some patients will return for SSC transplantation. This review summarizes the current knowledge and utility of SSC transplantation techniques. OBJECTIVE AND RATIONALE The aim of this narrative review is to provide an overview of the currently used experimental injection techniques for SSC transplantation in animal and human testes. This is crucial in understanding and determining the role of the different techniques necessary for successful transplantation. SEARCH METHODS A comprehensive review of peer-reviewed publications on this topic was performed using the PubMed and Google Scholar databases. The search was limited to English language work and studies between 1994 (from the first study on SSC transplantation) and April 2019. Key search terms included mouse, rat, boar, ram, dog, sheep, goat, cattle, monkey, human, cadaver, testes, SSC transplantation, injection and technique. OUTCOMES This review provides an extensive clinical overview of the current research in the field of human SSC transplantation. Rete testis injection with ultrasonography guidance currently seems the most promising injection technique thus far; however, the ability to draw clear conclusions is limited due to long ischemia time of cadaver testis, the relatively decreased volume of the testis, the diminishing size of seminiferous tubules, a lack of intratesticular pressure and leakage into the interstitium during the injection on human cadaver testis. Current evidence does not support improved outcomes from multiple infusions through the rete testes. Overall, further optimization is required to increase the efficiency and safety of the infusion method. WIDER IMPLICATIONS Identifying a favourable injection method for SSC transplantation will provide insight into the mechanisms of successful assisted human reproduction. Future research could focus on reducing leakage and establishing the optimal infusion cell concentrations and pressure.
Collapse
Affiliation(s)
- Murat Gul
- Laboratory of Reproductive Biology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark.,Department of Urology, Selcuk University School of Medicine, 42250 Konya, Turkey
| | - Simone Hildorf
- Department of Pediatric Surgery, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Lihua Dong
- Laboratory of Reproductive Biology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Jorgen Thorup
- Department of Pediatric Surgery, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Eva R Hoffmann
- DNRF Center for Chromosome Stability, Department of Molecular and Cellular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | | | - Jens Sønksen
- Department of Urology, Herlev and Gentofte University Hospital, 2930 Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Dina Cortes
- Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Department of Pediatrics, Copenhagen University Hospital Hvidovre, 2650 Hvidovre, Denmark
| | - Jens Fedder
- Centre of Andrology & Fertility Clinic, Department D, Odense University Hospital, 5000 Odense, Denmark.,Research Unit of Human Reproduction, Institute of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ellen Goossens
- Biology of the Testis, Research Laboratory for Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| |
Collapse
|
30
|
Liu H, Xie Y, Gao L, Sun X, Liang X, Deng C, Gao Y, Liu G. Impact on using cryopreservation of testicular or epididymal sperm upon intracytoplasmic sperm injection outcome in men with obstructive azoospermia: a systematic review and meta-analysis. J Assist Reprod Genet 2020; 37:2643-2651. [PMID: 32935172 DOI: 10.1007/s10815-020-01940-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/07/2020] [Indexed: 11/29/2022] Open
Abstract
PURPOSE To determine whether there was a significant impact on using cryopreservation of testicular or epididymal sperm upon the outcomes of intracytoplasmic sperm injection (ICSI) in patients with obstructive azoospermia (OA). METHOD Systematic review and meta-analysis of 20 retrospective studies in databases from January 1, 1995, to June 1, 2020. RESULT Twenty articles were included in this study. A total of 3602 (64.1%) of 5616 oocytes injected with fresh epididymal sperm were fertilized, compared with 2366 (61.2%) of 3862 oocytes injected with cryopreserved sperm (relative risk ratio (RR) 0.96, 95% confidence interval (CI) (0.90, 1.02), P > 0.05). A total of 303 (44.1%) of 687 ICSI cycles using fresh epididymal sperm resulted in a clinical pregnancy, compared with 150 (36.6%) of 410 ICSI cycles using cryopreserved epididymal sperm (RR 0.84, 95% CI (0.72, 0.97), P < 0.05). In the testis, a total of 2147 (68.7%) of 3125 oocytes injected with fresh sperm were fertilized, compared with 1623 (63.5%) of 2557 oocytes injected with cryopreserved sperm (RR 0.97, 95% CI (0.90, 1.06), P > 0.05). A total of 151 (47.8%) of 316 ICSI cycles using fresh testicular sperm resulted in a clinical pregnancy, compared with 113 (38.2%) of 296 ICSI cycles using cryopreserved sperm (RR 0.87, 95% CI (0.72, 1.05), P > 0.05). CONCLUSIONS In men with OA, there was a statistical lower clinical pregnancy rate (CPR) by using frozen epididymal sperm compared with fresh epididymal sperm, but showing no difference on fertilization rate (FR). Additionally, FR and CPR were not affected by whether the retrieved testicular sperm was frozen or fresh.
Collapse
Affiliation(s)
- Hanchao Liu
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yun Xie
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Linzhi Gao
- Reproductive Centre, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 58 Zhong shan 2nd Rd., Yuexiu District, Guangzhou, 510080, China
| | - Xiangzhou Sun
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Liang
- Reproductive Centre, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 58 Zhong shan 2nd Rd., Yuexiu District, Guangzhou, 510080, China
| | - Chunhua Deng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yong Gao
- Reproductive Medicine Center, The Key Laboratory for Reproductive Medicine of Guangdong Province, The First Affiliated Hospital of Sun Yat-sen University, No. 26 Yuan cun er heng Rd., Tianhe District, Guangzhou, China.
| | - Guihua Liu
- Reproductive Centre, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 58 Zhong shan 2nd Rd., Yuexiu District, Guangzhou, 510080, China.
| |
Collapse
|
31
|
Ferrari S, Paffoni A, Reschini M, Noli S, Dallagiovanna C, Guarneri C, Filippi F, Somigliana E. Variables affecting long-term usage rate of sperm samples cryopreserved for fertility preservation in cancer patients. Andrology 2020; 9:204-211. [PMID: 32814364 DOI: 10.1111/andr.12894] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/10/2020] [Accepted: 08/17/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Previous evidence highlighted that only a minority of men who banked their semen before cancer therapies subsequently used their frozen samples. This may question the economical validity of sperm cryopreservation programmes. However, in most contributions, the duration of follow-up was insufficient to draw robust information on the real rate of use. OBJECTIVES To shed more light on the potential benefits of cryopreservation programmes. MATERIALS AND METHODS Men who cryopreserved their semen in a public hospital for a diagnosis of cancer between 1986 and 2009 were retrospectively reviewed. The rate of use as well as the possible determinants was investigated. RESULTS The median time of follow-up was 12 [IQR: 7-16] years. One hundred forty-four patients out of 1,524 (9.4%, 95%CI: 8.1%-11.0%) used their frozen samples of whom 64% were azoospermic. The rate of men achieving parenthood with frozen semen was 46%. Predictive factors of use were older age at the time of storage, lower sperm count at the time of storage and a diagnosis of testicular cancer. The impact of this latter factor was also supported by the lower frequency of azoospermia after cancer treatment in these patients. DISCUSSION Cost-beneficial studies are warranted to assess and possibly improve the economical validity of sperm banking. CONCLUSION The usage rate of frozen sperm in cancer patient is low, even extending the duration of follow-up.
Collapse
Affiliation(s)
- Stefania Ferrari
- Obstetrics and Gynaecology Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Marco Reschini
- Obstetrics and Gynaecology Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Noli
- Obstetrics and Gynaecology Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Community and Clinical Sciences, University of Milan, Milan, Italy
| | - Chiara Dallagiovanna
- Obstetrics and Gynaecology Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Community and Clinical Sciences, University of Milan, Milan, Italy
| | - Cristina Guarneri
- Obstetrics and Gynaecology Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Filippi
- Obstetrics and Gynaecology Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Edgardo Somigliana
- Obstetrics and Gynaecology Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Community and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
32
|
Gomes FP, Park R, Viana AG, Fernandez-Costa C, Topper E, Kaya A, Memili E, Yates JR, Moura AA. Protein signatures of seminal plasma from bulls with contrasting frozen-thawed sperm viability. Sci Rep 2020; 10:14661. [PMID: 32887897 PMCID: PMC7474054 DOI: 10.1038/s41598-020-71015-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
The present study investigated the seminal plasma proteome of Holstein bulls with low (LF; n = 6) and high (HF; n = 8) sperm freezability. The percentage of viable frozen-thawed sperm (%ViableSperm) determined by flow cytometry varied from -2.2 in LF to + 7.8 in HF bulls, as compared to the average %ViableSperm (54.7%) measured in an 860-sire population. Seminal proteins were analyzed by label free mass spectrometry, with the support of statistical and bioinformatics analyses. This approach identified 1,445 proteins, associated with protein folding, cell-cell adhesion, NADH dehydrogenase activity, ATP-binding, proteasome complex, among other processes. There were 338 seminal proteins differentially expressed (p < 0.05) in LF and HF bulls. Based on multivariate analysis, BSP5 and seminal ribonuclease defined the HF phenotype, while spermadhesin-1, gelsolin, tubulins, glyceraldehyde-3-phosphate dehydrogenase, calmodulin, ATP synthase, sperm equatorial segment protein 1, peroxiredoxin-5, secretoglobin family 1D and glucose-6-phosphate isomerase characterized the LF phenotype. Regression models indicated that %ViableSperm of bulls was related to seminal plasma peroxiredoxin-5, spermadhesin-1 and the spermadhesin-1 × BSP5 interaction (R2 = 0.84 and 0.79; p < 0.05). This report is the largest dataset of bovine seminal plasma proteins. Specific proteins of the non-cellular microenvironment of semen are potential markers of sperm cryotolerance.
Collapse
Affiliation(s)
| | - Robin Park
- The Scripps Research Institute, La Jolla, CA, USA
| | | | | | | | | | | | - John R Yates
- The Scripps Research Institute, La Jolla, CA, USA.
| | | |
Collapse
|
33
|
A QUESTIONNAIRE SURVEY ON AWARENESS OF REPRODUCTIVE PROTECTION AND AUTOLOGOUS SPERM PRESERVATION AMONG CANCER PATIENTS. JOURNAL OF MEN'S HEALTH 2020. [DOI: 10.15586/jomh.v16i3.255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
34
|
Bourlon C, Riviello-Goya S, Acosta-Medina AA, Caballero-Landinez RE, Manrique-Rubio A, Teran-De-la-Sancha K, Gulias-Herrero A, Bourlon MT. Outcomes and Challenges of Reproductive Health in Hematopoietic Stem Cell Transplantation Survivors. Biol Blood Marrow Transplant 2020; 26:2127-2131. [PMID: 32659435 DOI: 10.1016/j.bbmt.2020.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 11/19/2022]
Abstract
Long-term therapy-related reproductive health side effects impact the quality of life of hematopoietic stem cell transplantation (HSCT) survivors. In this study, we evaluated the prevalence of gonadal dysfunction (GD) pre- and post-HSCT, analyzed factors associated with GD, and explored rates of fertility assessment (FA) and fertility preservation (FP) in a resource-limited setting. FA and outcomes of patients age ≤45 years undergoing HSCT between June 2000 and May 2018 were collected retrospectively. We included 213 patients with a median age of 26 years. Pre-HSCT FA was performed in 71.8%, with a GD rate of 17%. The rate of GD was not different between the sexes (females, 19.5% versus males, 16.1%; P = .616) and was only associated with increasing age. The rate of cryopreservation in the cohort was 3.3%. Almost one-half (47.7%) of post-HSCT patients completed FA and evidenced an increase in GD rate to 48.9%. Comparing pre-HSCT and post-HSCT GD rates, women had a significant increase (19.5% versus 81.4%; P < .001), whereas men did not (16.1% versus 20.4%; P = .76). These results were confirmed by a multiple imputation analysis accounting for missing data. Female sex, pre-HSCT cytotoxic therapy, myeloablative conditioning, and germ cell tumor (GCT) diagnosis were associated with post-HSCT GD. Reproductive health preservation can be positively impacted when FA and FP are prioritized at the initial diagnosis in HSCT candidates, particularly in women of older age and men with a diagnosis of GCT. The low FP success observed urges implementation of strategies that favor accessibility and improve quality of life of HSCT survivors in low- and middle-income countries.
Collapse
Affiliation(s)
- Christianne Bourlon
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Santiago Riviello-Goya
- Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Aldo A Acosta-Medina
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Rosa E Caballero-Landinez
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | | | - Alfonso Gulias-Herrero
- Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Maria T Bourlon
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
35
|
Gerstl B, Bertoldo MJ, Sullivan E, Volckmar X, Kerr A, Wand H, Ives A, Albalawi O, Anazodo A. Fatherhood Following Treatment for Testicular Cancer: A Systematic Review and Meta-Analyses. J Adolesc Young Adult Oncol 2020; 9:341-353. [DOI: 10.1089/jayao.2019.0164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Brigitte Gerstl
- Department of Biostatistics, The Kirby Institute, University of New South Wales, Sydney, Australia
- Kids Cancer Center, Sydney Children's Hospital, Sydney, Australia
| | - Michael J. Bertoldo
- School of Women's and Children's Health, University of New South Wales, Sydney, Australia
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Elizabeth Sullivan
- Faculty Health and Medicine, The University of Newcastle, Newcastle, Australia
| | - Xanthie Volckmar
- Department of Biostatistics, The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Aidan Kerr
- Department of Biostatistics, The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Handan Wand
- Department of Biostatistics, The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Angela Ives
- Cancer and Palliative Care Research and Evaluation Unit, University of Western Australia, Crawley, Australia
| | - Olayan Albalawi
- Department of Biostatistics, The Kirby Institute, University of New South Wales, Sydney, Australia
- Department of Statistics, Science College, Tabuk University, Tabuk, Saudi Arabia
| | - Antoinette Anazodo
- Kids Cancer Center, Sydney Children's Hospital, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales, Sydney, Australia
- Nelune Comprehensive Cancer Center, Prince of Wales Hospital, Sydney, Australia
| |
Collapse
|
36
|
Grin L, Girsh E, Harlev A. Male fertility preservation-Methods, indications and challenges. Andrologia 2020; 53:e13635. [PMID: 32390180 DOI: 10.1111/and.13635] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 12/23/2022] Open
Abstract
Male fertility preservation has been steadily increasing over the past two decades. Significant improvements have been achieved in the treatment modalities of cancer and other severe chronic medical conditions, leading to an increase in patient survivorship and the resulting demand for future parenthood. Recognition and proper patient counselling before commencing therapies with a potential gonadotoxic effect are of paramount importance. Similarly, nonmedically indicated fertility preservation is on the rise. Social sperm banking, gender dysphoria prior to affirmation procedures and posthumous reproduction preservation are becoming more common. When timing and logistics are appropriate, sperm cryopreservation is considered the gold standard for fertility preservation. Testicular tissue and spermatogonial stem cell autotransplantation is considered experimental and represents a promising alternative for pre-pubertal patients. The current paper aims to review the recent trends in male fertility preservation, the common indications for sperm cryopreservation, techniques for sperm retrieval and experimental frontiers.
Collapse
Affiliation(s)
- Leonti Grin
- Fertility and IVF Unit, Department of Obstetrics and Gynecology, Barzilai University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Eliezer Girsh
- Fertility and IVF Unit, Department of Obstetrics and Gynecology, Barzilai University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Avi Harlev
- Fertility and IVF Unit, Department of Obstetrics and Gynecology, Barzilai University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
37
|
Zhang H, Wang G, Cao M, Yin L, Xing Y, Wang J, Yang J, Zhang J. Level of Knowledge and Needs on Fertility Preservation in Reproductive-Aged Male Patients with Cancer. JOURNAL OF CANCER EDUCATION : THE OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER EDUCATION 2020; 35:321-326. [PMID: 30618008 PMCID: PMC7075821 DOI: 10.1007/s13187-018-1467-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
There is a growing concern about the fertility preservation (FP) for cancer patients of childbearing age. This study is the first in China to survey men with cancer, of reproductive age, regarding their knowledge of FP and their related needs. A 12-item cross-sectional survey was conducted of 332 male patients. The score for knowledge of FP was 3.5 ± 0.67, of a possible score of 8. Only 10.6% of the subjects had chosen to preserve fertility before treatments, but during therapy 68.7% wanted more information about FP. Younger patients were more likely have more knowledge concerning FP than older patients (odds ratio [OR] 0.86). The decision to make arrangements for FP before treatments was heavily influenced by being young and without children (OR, 0.78; OR, 0.11). Male cancer patients of reproductive age had limited knowledge of FP, and the majority was disinclined to make FP arrangements before therapy in China. Therefore, male cancer survivors should be well informed about FP soon after diagnosis and programs should be considered to improve the FP-related knowledge of male cancer survivors. We suggest that an assessment of patients' understanding of FP issues, before treatment, should be standard in clinical work.
Collapse
Affiliation(s)
- Hanfeng Zhang
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 55, 4th Section of Renmin South Road, Chengdu, 610040 Sichuan Province People’s Republic of China
| | - Guorong Wang
- Department of Nursing, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 55, 4th Section of Renmin South Road, Chengdu, 610040 Sichuan Province People’s Republic of China
| | - Maoqiu Cao
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 55, 4th Section of Renmin South Road, Chengdu, 610040 Sichuan Province People’s Republic of China
| | - Li Yin
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 55, 4th Section of Renmin South Road, Chengdu, 610040 Sichuan Province People’s Republic of China
| | - Yan Xing
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 55, 4th Section of Renmin South Road, Chengdu, 610040 Sichuan Province People’s Republic of China
| | - Jing Wang
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 55, 4th Section of Renmin South Road, Chengdu, 610040 Sichuan Province People’s Republic of China
| | - Jing Yang
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 55, 4th Section of Renmin South Road, Chengdu, 610040 Sichuan Province People’s Republic of China
| | - Jian Zhang
- Department of Nursing, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 55, 4th Section of Renmin South Road, Chengdu, 610040 Sichuan Province People’s Republic of China
| |
Collapse
|
38
|
Abstract
Spermatogonial stem cell (SSC) culture and transplantation pave the way for clinical restoration of fertility in male prepubertal cancer survivors. In this chapter we detail the steps for isolating and freezing testicular tissue along with protocols for the subsequent recovery from cryopreservation and transplantation of cells into a recipient testis. Transplantation of cultured or thawed SSCs provides not only a functional assay for identification of stem cells, a critical tool for the study of the germline stem cell niche in model organisms, but also a framework for reconstitution of spermatogenesis in humans. As proof of concept, the outlined methods have been performed successfully in the murine model and have the potential to be translated to clinical environments.
Collapse
|
39
|
Yuan Y, Li L, Cheng Q, Diao F, Zeng Q, Yang X, Wu Y, Zhang H, Huang M, Chen J, Zhou Q, Zhu Y, Hua R, Tian J, Wang X, Zhou Z, Hao J, Yu J, Hua D, Liu J, Guo X, Zhou Q, Sha J. In vitro testicular organogenesis from human fetal gonads produces fertilization-competent spermatids. Cell Res 2020; 30:244-255. [PMID: 32086476 DOI: 10.1038/s41422-020-0283-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 02/03/2020] [Indexed: 01/18/2023] Open
Abstract
Unlike most organs that mature during the fetal period, the male reproductive system reaches maturity only at puberty with the commencement of spermatogenesis. Robust modelling of human testicular organogenesis in vitro would facilitate research into mechanisms of and factors affecting human spermatogenic failure and male fertility preservation in prepubertal tumor patients. Here, we report successful recapitulation of human testicular organogenesis in vitro from fetal gonadal ridge. Our model displayed the formation of mature seminiferous epithelium and self-renewing spermatogonia. Remarkably, in vitro-derived haploid spermatids have undergone meiotic recombination, and showed increased genetic diversity as indicated by genetic analysis. Moreover, these spermatids were able to fertilize oocytes and support subsequent blastocyst formation. The in vitro testicular organogenesis system described here will play an important role in elucidating the regulation of human testis development and maintaining male fertility in prepubertal cancer patients.
Collapse
Affiliation(s)
- Yan Yuan
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, 214062, Wuxi, Jiangsu, China.,State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Laihua Li
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Qing Cheng
- State Key Laboratory of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Medical University, 210029, Nanjing, Jiangsu, China
| | - Feiyang Diao
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, 210029, Nanjing, Jiangsu, China
| | - Qiao Zeng
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, 210029, Nanjing, Jiangsu, China
| | - Xiaoyu Yang
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, 210029, Nanjing, Jiangsu, China
| | - Yibo Wu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, 214062, Wuxi, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Mingqian Huang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Junqing Chen
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Quan Zhou
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Yunfei Zhu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Rong Hua
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Jianyu Tian
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Xin Wang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Zuomin Zhou
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Jie Hao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Jinjin Yu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, 214062, Wuxi, Jiangsu, China
| | - Dong Hua
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, 214062, Wuxi, Jiangsu, China
| | - Jiayin Liu
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, 210029, Nanjing, Jiangsu, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China.
| | - Qi Zhou
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, Jiangsu, China. .,State Key Laboratory of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Medical University, 210029, Nanjing, Jiangsu, China.
| |
Collapse
|
40
|
Levi-Setti PE, Negri L, Baggiani A, Morenghi E, Albani E, Dioguardi CMC, Specchia C, Patrizio P. Testicular sperm extraction and intracytoplasmic sperm injection outcome in cancer survivors with no available cryopreserved sperm. J Assist Reprod Genet 2020; 37:875-882. [PMID: 31981037 DOI: 10.1007/s10815-020-01697-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 01/17/2020] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE To assess rates of successful testicular sperm retrieval and intracytoplasmic sperm injection (ICSI) outcome in cancer survivors affected by non-obstructive azoospermia (NOA) or retrograde ejaculation (RE)/failure of emission (FOE). METHODS A retrospective analysis of cancer survivors who did not cryopreserve sperm prior to treatment undergoing testicular sperm extraction (TESE). Non-cancer NOA patients and neurologic RE/FOE were the control group. RESULTS A total of 97 cancer survivors were offered TESE and 88 (91%) accepted. Sperm was retrieved and cryopreserved in 34/67 patients with NOA (50.7%) and in 21/21 patients affected by RE/FOE (100%). Sperm retrieval rates were similar in the control group (44.9% in NOA and 100% in RE/FOE). The ICSI cumulative pregnancy rate (60%) and live birth rate (40%) per couple in 30 NOA men did not differ from controls (50.0 and 46.5%, respectively; p = 0.399/0.670). The cumulative pregnancy rate (66.7%) and live birth rate (55.6%) in 18 RE/FOE men did not differ from the control group (38.9 and 33.3%, respectively; p = 0.181/0.315). The cancer type and the resulting infertility disorder (NOA or RE/FOE) were not associated with ICSI outcomes. Female partner age was inversely related to the cumulative live birth rate, being fourfold lower (11.5%) in women ≥ 40 years and 48.8% in younger women (p = 0.0037). CONCLUSIONS The rate of successful TESE and the ICSI outcome in cancer survivors with NOA and RE/FOE is the same as non-cancer azoospermic patients. Female partner age (older than 40 years) was associated with a significant reduction in live birth rates after TESE-ICSI procedures.
Collapse
Affiliation(s)
- Paolo Emanuele Levi-Setti
- Humanitas Fertility Centre, Department of Gynecology, Division of Gynecology and Reproductive Medicine, Humanitas Clinical and Research Center - IRCCS, via Manzoni 56, 20089, Rozzano, Milan, Italy.
- Department of Obstetrics, Gynaecology and Reproductive Sciences, Yale University, School of Medicine, New Haven, CT, USA.
| | - Luciano Negri
- Humanitas Fertility Centre, Department of Gynecology, Division of Gynecology and Reproductive Medicine, Humanitas Clinical and Research Center - IRCCS, via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Annamaria Baggiani
- Humanitas Fertility Centre, Department of Gynecology, Division of Gynecology and Reproductive Medicine, Humanitas Clinical and Research Center - IRCCS, via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Emanuela Morenghi
- Biostatistics Unit, Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Elena Albani
- Humanitas Fertility Centre, Department of Gynecology, Division of Gynecology and Reproductive Medicine, Humanitas Clinical and Research Center - IRCCS, via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Carola Maria Conca Dioguardi
- Humanitas Fertility Centre, Department of Gynecology, Division of Gynecology and Reproductive Medicine, Humanitas Clinical and Research Center - IRCCS, via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Cristina Specchia
- Humanitas Fertility Centre, Department of Gynecology, Division of Gynecology and Reproductive Medicine, Humanitas Clinical and Research Center - IRCCS, via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Pasquale Patrizio
- Department of Obstetrics, Gynaecology and Reproductive Sciences, Yale University, School of Medicine, New Haven, CT, USA
| |
Collapse
|
41
|
Fu L, An Q, Zhang K, Liu Y, Tong Y, Xu J, Zhou F, Wang X, Guo Y, Lu W, Liang X, Gu Y. Quantitative proteomic characterization of human sperm cryopreservation: using data-independent acquisition mass spectrometry. BMC Urol 2019; 19:133. [PMID: 31842847 PMCID: PMC6916233 DOI: 10.1186/s12894-019-0565-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 12/02/2019] [Indexed: 01/09/2023] Open
Abstract
Background Human sperm cryopreservation is a simple and effective approach for male fertility preservation. Methods To identify potential proteomic changes in this process, data-independent acquisition (DIA), a technology with high quantitative accuracy and highly reproducible proteomics, was used to quantitatively characterize the proteomics of human sperm cryopreservation. Results A total of 174 significantly differential proteins were identified between fresh and cryoperservated sperm: 98 proteins decreased and 76 proteins increased in the cryopreservation group. Bioinformatic analysis revealed that metabolic pathways play an important role in cryopreservation, including: propanoate metabolism, glyoxylate and dicarboxylate metabolism, glycolysis/gluconeogenesis, and pyruvate metabolism. Four different proteins involved in glycolysis were identified by Western blotting: GPI, LDHB, ADH5, and PGAM1. Conclusions Our work will provide valuable information for future investigations and pathological studies involving sperm cryopreservation.
Collapse
Affiliation(s)
- Longlong Fu
- National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, 100081, China.,Department of Male Clinical Research/Human sperm bank, National Research Institute for Family Planning & WHO Collaborating Center for Research in Human Reproduction, Beijing, 100081, China
| | - Qi An
- National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, 100081, China.,Department of Male Clinical Research/Human sperm bank, National Research Institute for Family Planning & WHO Collaborating Center for Research in Human Reproduction, Beijing, 100081, China.,Graduate School of Peking Union Medical College, Beijing, 100730, China
| | - Kaishu Zhang
- Department of Reproductive Medicine, The Afliated Hospital of Qingdao University, Qingdao, Shandong, 266000, People's Republic of China
| | - Ying Liu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, 215025, China
| | - Yue Tong
- National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, 100081, China.,Department of Male Clinical Research/Human sperm bank, National Research Institute for Family Planning & WHO Collaborating Center for Research in Human Reproduction, Beijing, 100081, China.,Graduate School of Peking Union Medical College, Beijing, 100730, China
| | - Jianfeng Xu
- National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, 100081, China.,Department of Male Clinical Research/Human sperm bank, National Research Institute for Family Planning & WHO Collaborating Center for Research in Human Reproduction, Beijing, 100081, China
| | - Fang Zhou
- National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, 100081, China.,Department of Male Clinical Research/Human sperm bank, National Research Institute for Family Planning & WHO Collaborating Center for Research in Human Reproduction, Beijing, 100081, China
| | - Xiaowei Wang
- National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, 100081, China.,Department of Male Clinical Research/Human sperm bank, National Research Institute for Family Planning & WHO Collaborating Center for Research in Human Reproduction, Beijing, 100081, China
| | - Ying Guo
- National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, 100081, China.,Department of Male Clinical Research/Human sperm bank, National Research Institute for Family Planning & WHO Collaborating Center for Research in Human Reproduction, Beijing, 100081, China
| | - Wenhong Lu
- National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, 100081, China. .,Graduate School of Peking Union Medical College, Beijing, 100730, China.
| | - Xiaowei Liang
- Department of Male Clinical Research/Human sperm bank, National Research Institute for Family Planning & WHO Collaborating Center for Research in Human Reproduction, Beijing, 100081, China
| | - Yiqun Gu
- National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, 100081, China. .,Department of Male Clinical Research/Human sperm bank, National Research Institute for Family Planning & WHO Collaborating Center for Research in Human Reproduction, Beijing, 100081, China. .,Graduate School of Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
42
|
Dong L, Gul M, Hildorf S, Pors SE, Kristensen SG, Hoffmann ER, Cortes D, Thorup J, Andersen CY. Xeno-Free Propagation of Spermatogonial Stem Cells from Infant Boys. Int J Mol Sci 2019; 20:ijms20215390. [PMID: 31671863 PMCID: PMC6862004 DOI: 10.3390/ijms20215390] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/03/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022] Open
Abstract
Spermatogonial stem cell (SSC) transplantation therapy is a promising strategy to renew spermatogenesis for prepubertal boys whose fertility is compromised. However, propagation of SSCs is required due to a limited number of SSCs in cryopreserved testicular tissue. This propagation must be done under xeno-free conditions for clinical application. SSCs were propagated from infant testicular tissue (7 mg and 10 mg) from two boys under xeno-free conditions using human platelet lysate and nutrient source. We verified SSC-like cell clusters (SSCLCs) by quantitative real-time polymerase chain reaction (PCR) and immune-reaction assay using the SSC markers undifferentiated embryonic cell transcription factor 1 (UTF1), ubiquitin carboxyl-terminal hydrolase isozyme L1 (UCHL1), GDNF receptor alpha-1 (GFRα-1) Fα and promyelocytic leukaemia zinc finger protein (PLZF). The functionality of the propagated SSCs was investigated by pre-labelling using green fluorescent Cell Linker PKH67 and xeno-transplantation of the SSCLCs into busulfan-treated, therefore sterile, immunodeficient mice. SSC-like cell clusters (SSCLCs) appeared after 2 weeks in primary passage. The SSCLCs were SSC-like as the UTF1, UCHL1, GFRα1 and PLZF were all positive. After 2.5 months’ culture period, a total of 13 million cells from one sample were harvested for xenotransplantation. Labelled human propagated SSCs were identified and verified in mouse seminiferous tubules at 3–6 weeks, confirming that the transplanted cells contain SSCLCs. The present xeno-free clinical culture protocol allows propagation of SSCs from infant boys.
Collapse
Affiliation(s)
- Lihua Dong
- Laboratory of Reproductive Biology, Rigshospitalet, University Hospital of Copenhagen, 2100 Copenhagen, Denmark.
| | - Murat Gul
- Laboratory of Reproductive Biology, Rigshospitalet, University Hospital of Copenhagen, 2100 Copenhagen, Denmark.
- Department of Urology, Aksaray University School of Medicine, Aksaray 68100, Turkey.
| | - Simone Hildorf
- Department of Pediatric Surgery, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark.
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, Rigshospitalet, University Hospital of Copenhagen, 2100 Copenhagen, Denmark.
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, Rigshospitalet, University Hospital of Copenhagen, 2100 Copenhagen, Denmark.
| | - Eva R Hoffmann
- Center for Chromosome Stability, Institute of Molecular and Cellular Medicine, 2200 Copenhagen, Denmark.
- Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Dina Cortes
- Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
- Department of Pediatrics, Hvidovre, Copenhagen University Hospital, 2650 Copenhagen, Denmark.
| | - Jorgen Thorup
- Department of Pediatric Surgery, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark.
- Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Rigshospitalet, University Hospital of Copenhagen, 2100 Copenhagen, Denmark.
- Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
43
|
Fertility preservation in patients with hematologic malignancies and recipients of hematopoietic cell transplants. Blood 2019; 134:746-760. [PMID: 31292116 DOI: 10.1182/blood.2018846790] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 05/30/2019] [Indexed: 02/07/2023] Open
Abstract
Patients with hematologic malignancies and those undergoing hematopoietic cell transplantation (HCT) face a complex set of challenges when considering options for fertility preservation (FP). There are no standard options for prepubertal children, and women with hematologic malignancies may not be eligible for standard FP options. Fortunately, initial therapies for most blood cancers are not highly gonadotoxic, affording an important opportunity for postremission counseling and referrals to fertility specialists. These patients face a high risk of relapse, and many will be referred for autologous or allogeneic HCT, which carries an extremely high risk of infertility. The expanding indications for HCT to include benign hematologic disorders as well as autoimmune diseases mandate that all hematologists are familiar with these risks. Oncofertility researchers are continually pushing the boundaries of what may be possible for our patients; in the meantime, communication and shared decision-making between hematologists and patients, as well as program-building, education, and outreach are essential to ensure that these patients, many of whom will be cured, maintain all of their options for a fulfilling life after intensive therapy.
Collapse
|
44
|
Gebauer J, Higham C, Langer T, Denzer C, Brabant G. Long-Term Endocrine and Metabolic Consequences of Cancer Treatment: A Systematic Review. Endocr Rev 2019; 40:711-767. [PMID: 30476004 DOI: 10.1210/er.2018-00092] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/21/2018] [Indexed: 02/08/2023]
Abstract
The number of patients surviving ≥5 years after initial cancer diagnosis has significantly increased during the last decades due to considerable improvements in the treatment of many cancer entities. A negative consequence of this is that the emergence of long-term sequelae and endocrine disorders account for a high proportion of these. These late effects can occur decades after cancer treatment and affect up to 50% of childhood cancer survivors. Multiple predisposing factors for endocrine late effects have been identified, including radiation, sex, and age at the time of diagnosis. A systematic literature search has been conducted using the PubMed database to offer a detailed overview of the spectrum of late endocrine disorders following oncological treatment. Most data are based on late effects of treatment in former childhood cancer patients for whom specific guidelines and recommendations already exist, whereas current knowledge concerning late effects in adult-onset cancer survivors is much less clear. Endocrine sequelae of cancer therapy include functional alterations in hypothalamic-pituitary, thyroid, parathyroid, adrenal, and gonadal regulation as well as bone and metabolic complications. Surgery, radiotherapy, chemotherapy, and immunotherapy all contribute to these sequelae. Following irradiation, endocrine organs such as the thyroid are also at risk for subsequent malignancies. Although diagnosis and management of functional and neoplastic long-term consequences of cancer therapy are comparable to other causes of endocrine disorders, cancer survivors need individually structured follow-up care in specialized surveillance centers to improve care for this rapidly growing group of patients.
Collapse
Affiliation(s)
- Judith Gebauer
- Experimental and Clinical Endocrinology, University Hospital of Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Claire Higham
- Department of Endocrinology, Christie Hospital NHS Foundation Trust, Manchester, United Kingdom.,Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Thorsten Langer
- Division of Pediatric Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Christian Denzer
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Endocrinology and Diabetes, Ulm University Medical Center, Ulm, Germany
| | - Georg Brabant
- Experimental and Clinical Endocrinology, University Hospital of Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Department of Endocrinology, Christie Hospital NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
45
|
Seminiferous tubule molecular imaging for evaluation of male fertility: Seeing is believing. Tissue Cell 2019; 58:24-32. [PMID: 31133243 DOI: 10.1016/j.tice.2019.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/28/2019] [Accepted: 04/05/2019] [Indexed: 01/15/2023]
Abstract
The proper assessment of male fertility is essential for diagnosing and treating male infertility. Currently, spermiogram and Johnsen testicular biopsy score counts are used to assess male fertility. However, spermiogram is not a suitable option for non-obstructive azoospermia patients, and Johnsen testicular biopsy scores only represent localized and not the overall spermatogenesis. Whole-mount staining was a novel method for evaluating protein expression in the tissue. Thus, we explored its application in human seminiferous tubules. Testicular biopsies from 57 azoospermia patients were categorized as obstructive azoospermia (OA), maturation arrest (MA) and Sertoli-cells only syndrome (SCOS). We performed whole-mount staining of their seminiferous tubules and evaluated the spermatogonial stem cells (SSCs), differentiated spermatogonia (SG), spermatocytes (SPC) and spermatids (SD) with their respective markers (GFRA1, CD117, SYCP3, and PNA) to assess fertility. GFRA1, CD117, SYCP3, and PNA were not expressed in SCOS patients, whereas all of them were detected in OA patients. In MA patients with arrested spermatogenesis at the SPC stage, GFRA1, CD117, and SYCP3, but not PNA were expressed in the seminiferous tubules. In MA patients with arrested spermatogenesis at the spermatogonia stage, only GFRA1 was expressed in the seminiferous tubules. These results were consistent with the Johnsen testicular biopsy score counts except for one patient, where although only Sertoli cells were indicated by the score, SSCs were also detected in the whole-mounts. Collectively, whole-mount staining could be used to analyze the inherent spermatogenesis of seminiferous tubules through staining of germ cells at different stages. It offers a more accurate and promising faster method for assessing male fertility compared with traditional biopsy screening. And it could have potential value for the clinical purpose for male fertility management.
Collapse
|
46
|
Jia Y, Lue Y, Swerdloff RS, Lasky JL, Panosyan EH, Dai-Ju J, Wang C. The humanin analogue (HNG) prevents temozolomide-induced male germ cell apoptosis and other adverse effects in severe combined immuno-deficiency (SCID) mice bearing human medulloblastoma. Exp Mol Pathol 2019; 109:42-50. [PMID: 31085184 DOI: 10.1016/j.yexmp.2019.104261] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 05/10/2019] [Indexed: 02/05/2023]
Abstract
Subfertility is a major concern of long-term cancer survivors at the reproductive age. We have previously demonstrated that a potent humanin analogue, HNG, protected chemotherapy-induced apoptosis in germ cells but not cancer cells in a metastatic melanoma allograft model. In this study, we utilized severe combined immuno-deficiency (SCID) mice bearing human medulloblastoma to study the effect of HNG in Temozolomide (TMZ) induced male germ cell apoptosis and white blood cell (WBC) suppression. Human medulloblastoma DAOY cells were injected subcutaneously into the right flank of male SCID mice. Three weeks later, groups of tumor-bearing mice received one of the following treatments: vehicle, HNG, TMZ, or TMZ + HNG. 24 h after last injection, the tumors weights, complete blood counts, liver and spleen weights, male germ cell apoptosis was assessed. HNG did not affect TMZ's significant anti-tumor action. HNG significantly prevented TMZ-induced germ cell apoptosis and attenuated the suppressed total WBC and granulocyte counts in SCID mice with or without TMZ treatment. HNG also attenuated TMZ-induced body weight loss and decrease of spleen and liver weights. In conclusion, HNG ameliorated TMZ-induced germ cell apoptosis; WBC and granulocytes loss; and decreased body/organ weights without compromising the TMZ's anti-cancer action on medulloblastoma xenografts in SCID mice.
Collapse
Affiliation(s)
- Yue Jia
- Division of Endocrinology, Department of Medicine, Los Angeles Biomedical Research Institute and Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Yanhe Lue
- Division of Endocrinology, Department of Medicine, Los Angeles Biomedical Research Institute and Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Ronald S Swerdloff
- Division of Endocrinology, Department of Medicine, Los Angeles Biomedical Research Institute and Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Joseph L Lasky
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Los Angeles Biomedical Research Institute and Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Eduard H Panosyan
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Los Angeles Biomedical Research Institute and Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Jenny Dai-Ju
- Division of Endocrinology, Department of Medicine, Los Angeles Biomedical Research Institute and Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Christina Wang
- Division of Endocrinology, Department of Medicine, Los Angeles Biomedical Research Institute and Harbor-UCLA Medical Center, Torrance, CA, United States of America.
| |
Collapse
|
47
|
Fu L, Zhou F, An Q, Zhang K, Wang X, Xu J, Guo Y, Lu W, Liang X, Gu Y. Sperm Cryopreservation for Male Cancer Patients: More than 10 Years of Experience, in Beijing China. Med Sci Monit 2019; 25:3256-3261. [PMID: 31048670 PMCID: PMC6511111 DOI: 10.12659/msm.913513] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Fertility preservation is very important for male cancer patients, especially adolescents. Unfortunately, the use of fertility preservation is very low among Chinese male cancer patients. Additionally, the cumulative rate of frozen sperm use is also low. Material/Methods We performed a retrospective study by collecting available information at the Human Sperm Bank, National Research Institute for Family Planning from July 2006 to December 2017 to examine the data in China. Results A total 145 male cancer patients underwent sperm cryopreservation. The patients were 29.3±6.9 years old, and 6.2% (9 out of 145) of the patients were adolescents under the age of 18 years old. As of June 2018, only 9.7% (14 out of 145) of patients returned to use their cryopreserved sperm for assisted reproduction technology (ART). Of the 33 ART cycles, conceptions were achieved in 51.5% (17 out of 33), and the rate of patients who had a baby was 71.4% (10 out of 14). The data indicate men with testicular cancer or leukemia had lower total sperm counts and recovery rate of progressive sperm than did men with other types of cancer, while men with sarcoma had the lowest progressive sperm. Conclusions The physician should make an effort to promote fertility preservation for male cancer patients in China. And patients with testicular cancer and leukemia require additional attention.
Collapse
Affiliation(s)
- Longlong Fu
- Department of Male Clinical Research/Human Sperm Bank, National Research Institute for Family Planning, and WHO Collaborating Centre for Research in Human Reproduction, Beijing, China (mainland).,Graduate School of Peking Union Medical College, Beijing, China (mainland).,National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, China (mainland)
| | - Fang Zhou
- Department of Male Clinical Research/Human Sperm Bank, National Research Institute for Family Planning, and WHO Collaborating Centre for Research in Human Reproduction, Beijing, China (mainland).,National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, China (mainland)
| | - Qi An
- Department of Male Clinical Research/Human Sperm Bank, National Research Institute for Family Planning, and WHO Collaborating Centre for Research in Human Reproduction, Beijing, China (mainland).,Graduate School of Peking Union Medical College, Beijing, China (mainland).,National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, China (mainland)
| | - Kaishu Zhang
- Department of Reproductive Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Xiaowei Wang
- Department of Male Clinical Research/Human Sperm Bank, National Research Institute for Family Planning, and WHO Collaborating Centre for Research in Human Reproduction, Beijing, China (mainland).,National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, China (mainland)
| | - Jianfeng Xu
- Department of Male Clinical Research/Human Sperm Bank, National Research Institute for Family Planning, and WHO Collaborating Centre for Research in Human Reproduction, Beijing, China (mainland).,National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, China (mainland)
| | - Ying Guo
- Department of Male Clinical Research/Human Sperm Bank, National Research Institute for Family Planning, and WHO Collaborating Centre for Research in Human Reproduction, Beijing, China (mainland).,National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, China (mainland)
| | - Wenhong Lu
- Department of Male Clinical Research/Human Sperm Bank, National Research Institute for Family Planning, and WHO Collaborating Centre for Research in Human Reproduction, Beijing, China (mainland).,National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, China (mainland)
| | - Xiaowei Liang
- Department of Male Clinical Research/Human Sperm Bank, National Research Institute for Family Planning, and WHO Collaborating Centre for Research in Human Reproduction, Beijing, China (mainland)
| | - Yiqun Gu
- Department of Male Clinical Research/Human Sperm Bank, National Research Institute for Family Planning, and WHO Collaborating Centre for Research in Human Reproduction, Beijing, China (mainland).,Graduate School of Peking Union Medical College, Beijing, China (mainland).,National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, China (mainland)
| |
Collapse
|
48
|
Baetas J, Rabaça A, Gonçalves A, Barros A, Sousa M, Sá R. Protective role of N-acetylcysteine (NAC) on human sperm exposed to etoposide. Basic Clin Androl 2019; 29:3. [PMID: 30774957 PMCID: PMC6366041 DOI: 10.1186/s12610-018-0082-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/28/2018] [Indexed: 01/02/2023] Open
Abstract
Background Although recent progress in cancer treatment has increased patient survival and improved quality of life, reproductive side effects are still for concern. One way to decrease gonadal impairment is to use cytoprotectors. In testicular cancer, etoposide is generally used in combination with other agents, but there are no in-vitro studies of sperm exposure to etoposide and cytoprotectors, namely N-acetylcysteine (NAC). Methods Twenty semen samples were individually divided into five groups: control, incubation with NAC alone, incubation with etoposide alone, sequential exposure of NAC followed by etoposide (pre-treatment) and sequential exposure of etoposide followed by NAC (post-treatment). Sperm characteristics, chromatin condensation (aniline blue), DNA fragmentation (TUNEL), oxidative stress (OxyDNA labelling) and glutathione quantification were used to evaluate the capabilities of NAC as a prophylactic (pre-treatment) or ameliorator (post-treatment) agent over the effects caused in sperm during in-vitro exposure to etoposide. Results No deleterious effects were observed on sperm motility or sperm membrane integrity. Results revealed that prophylactic use of NAC (pre-treatment) increased rates of immature sperm chromatin as compared to ameliorator use of NAC (post-treatment), and increased rates of sperm DNA fragmentation in relation to controls. Pre and post-treatment with NAC increased oxidative levels in comparison to controls, but also increased levels of cellular antioxidant glutathione. Conclusions The results indicate that NAC has the ability to counteract etoposide-induced toxicity rather than preventing the etoposide cytotoxic effects over sperm DNA, suggesting that the administration of NAC to cells formerly exposed to etoposide is preferable to its prophylactic use. As the results evidenced that NAC seems to be more efficient in attenuating sperm etoposide cytotoxic effects instead of being used as a chemoprophylactic agent, this reinforces the idea that there might be a new NAC mechanism over DNA.
Collapse
Affiliation(s)
- João Baetas
- 1Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.,2Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Ana Rabaça
- 1Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Ana Gonçalves
- Centre for Reproductive Genetics A. Barros (CGR), Av. do Bessa, 240, 1° Dto. Frente, 4100-012 Porto, Portugal
| | - Alberto Barros
- Centre for Reproductive Genetics A. Barros (CGR), Av. do Bessa, 240, 1° Dto. Frente, 4100-012 Porto, Portugal.,4Department of Genetics, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.,5Health Institute of Research and Innovation (IPATIMUP/i3S), University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Mário Sousa
- 1Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.,6Multidisciplinary Unit for Biomedical Research (UMIB), University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Rosália Sá
- 1Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.,6Multidisciplinary Unit for Biomedical Research (UMIB), University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
49
|
Cwynarski K, Marzolini MAV, Barrington SF, Follows G, Illidge T, Stern S, Davies A. The management of primary mediastinal B‐cell lymphoma: a British Society for Haematology Good Practice Paper. Br J Haematol 2019; 185:402-409. [DOI: 10.1111/bjh.15731] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Kate Cwynarski
- Department of Haematology University College London Hospitals NHS Foundation TrustLondonUK
| | - Maria A. V. Marzolini
- Department of Haematology University College London Hospitals NHS Foundation TrustLondonUK
| | - Sally F. Barrington
- King's College London and Guy's and St Thomas’ PET Centre School of Biomedical Engineering and Imaging Sciences King's College London King's Health Partners LondonUK
| | - George Follows
- Department of Haematology Addenbrooke's Hospital CambridgeUK
| | - Timothy Illidge
- Department of Clinical Oncology Christie Hospital ManchesterUK
| | - Simon Stern
- Department of Haematology St Helier Hospital CarshaltonUK
| | - Andrew Davies
- Department of Medical Oncology Southampton General Hospital Southampton UK
| | | |
Collapse
|
50
|
Rezaei Topraggaleh T, Rezazadeh Valojerdi M, Montazeri L, Baharvand H. A testis-derived macroporous 3D scaffold as a platform for the generation of mouse testicular organoids. Biomater Sci 2019; 7:1422-1436. [DOI: 10.1039/c8bm01001c] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Extracellular matrix-derived scaffolds provide an efficient platform for the generation of organ-like structures.
Collapse
Affiliation(s)
| | | | - Leila Montazeri
- Department of Cell Engineering
- Cell Science Research Center
- Royan Institute for Stem Cell Biology and Technology
- ACECR
- Tehran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology at Cell Science Research Center
- Royan Institute for Stem Cell Biology and Technology
- ACECR
- Tehran
- Iran
| |
Collapse
|