1
|
Rambhatla A, Shah R, Ziouziou I, Kothari P, Salvio G, Gul M, Hamoda T, Kavoussi P, Atmoko W, Toprak T, Birowo P, Ko E, Arafa M, Ghayda RA, Karthikeyan VS, Russo GI, Pinggera GM, Chung E, Harraz AM, Martinez M, Phuoc NHV, Tadros N, Saleh R, Savira M, Colpi GM, Zohdy W, Pescatori E, Park HJ, Fukuhara S, Tsujimura A, Rojas-Cruz C, Marino A, Mak SK, Amar E, Ibrahim W, Sindhwani P, Alhathal N, Busetto GM, Al Hashimi M, El-Sakka A, Ramazan A, Dimitriadis F, Timpano M, Jezek D, Altay B, Zylbersztejn DS, Wong MYC, Moon DG, Wyns C, Gamidov S, Akhavizadegan H, Franceschelli A, Aydos K, Quang N, Ashour S, Al Dayel A, Al-Marhoon MS, Micic S, Binsaleh S, Hussein A, Elbardisi H, Mostafa T, Ramsay J, Zachariou A, Abdelrahman IFS, Rajmil O, Kalkanli A, Molina JMC, Bocu K, Duarsa GWK, Çeker G, Serefoglu EC, Bahar F, Gherabi N, Kuroda S, Bouzouita A, Gudeloglu A, Ceyhan E, Hasan MSM, Musa MU, Motawi A, Cho CL, Taniguchi H, Ho CCK, Vazquez JFS, Mutambirwa S, Gungor ND, Bendayan M, Giulioni C, Baser A, Falcone M, Boeri L, Blecher G, Kheradmand A, Sethupathy T, Adriansjah R, Narimani N, Konstantinidis C, Nguyen TT, Japari A, Dolati P, Singh K, Ozer C, Sarikaya S, Sheibak N, Bosco NJ, Özkent MS, Le ST, Sokolakis I, Katz D, Smith R, Truong MN, Le TV, Huang Z, Deger MD, Arslan U, Calik G, Franco G, Rashed A, Kahraman O, Andreadakis S, Putra R, Balercia G, Khalafalla K, Cannarella R, Tuấn AĐ, El Meliegy A, Zilaitiene B, Ramirez MLZ, Giacone F, Calogero AE, Makarounis K, Jindal S, Hoai BN, Banthia R, Peña MR, Moorthy D, Adamyan A, Kulaksiz D, Kandil H, Sofikitis N, Salzano C, Jungwirth A, Banka SR, Mierzwa TC, Turunç T, Jain D, Avoyan A, Salacone P, Kadıoğlu A, Gupta C, Lin H, Shamohammadi I, Mogharabian N, Barrett T, Danacıoğlu YO, Crafa A, Daoud S, Malhotra V, Almardawi A, Selim OM, Moussa M, Haghdani S, Duran MB, Kunz Y, Preto M, Eugeni E, Nguyen T, Elshahid AR, Suyono SS, Parikesit D, Nada E, Orozco EG, Boitrelle F, Trang NTM, Jamali M, Nair R, Ruzaev M, Gadda F, Thomas C, Ferreira RH, Gul U, Maruccia S, Kanbur A, Kinzikeeva E, Abumelha SM, Kosgi R, Gokalp F, Soebadi MA, Paul GM, Sajadi H, Gupte D, Ambar RF, Sogutdelen E, Singla K, Basukarno A, Kim SHK, Gilani MAS, Nagao K, Brodjonegoro SR, Rezano A, Elkhouly M, Mazzilli R, Farsi HMA, Ba HN, Alali H, Kafetzis D, Long TQT, Alsaid S, Cuong HBN, Oleksandr K, Mustafa A, Acosta H, Pai H, Şahin B, Arianto E, Teo C, Jayaprakash SP, Rachman RI, Yenice MG, Sefrioui O, Priyadarshi S, Tanic M, Alfatlaw NK, Rizaldi F, Vishwakarma RB, Kanakis G, Cherian DT, Lee J, Galstyan R, Keskin H, Wurzacher J, Seno DH, Noegroho BS, Margiana R, Javed Q, Castiglioni F, Tanwar R, Puigvert A, Kaya C, Purnomo M, Yazbeck C, Amir A, Borges E, Bellavia M, Deswanto IA, KV V, Liguori G, Minh DH, Siddiqi K, Colombo F, Zini A, Patel N, Çayan S, Al-kawaz U, Ragab M, Hebrard GH, de la Rosette J, Efesoy O, Hoffmann I, Teixeira TA, Saylam B, Delgadillo D, Agarwal A. Global Practice Patterns and Variations in the Medical and Surgical Management of Non-Obstructive Azoospermia: Results of a World-Wide Survey, Guidelines and Expert Recommendations. World J Mens Health 2025; 43:92-122. [PMID: 38606867 PMCID: PMC11704169 DOI: 10.5534/wjmh.230339] [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: 11/22/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 04/13/2024] Open
Abstract
PURPOSE Non-obstructive azoospermia (NOA) is a common, but complex problem, with multiple therapeutic options and a lack of clear guidelines. Hence, there is considerable controversy and marked variation in the management of NOA. This survey evaluates contemporary global practices related to medical and surgical management for patients with NOA. MATERIALS AND METHODS A 56-question online survey covering various aspects of the evaluation and management of NOA was sent to specialists around the globe. This paper analyzes the results of the second half of the survey dealing with the management of NOA. Results have been compared to current guidelines, and expert recommendations have been provided using a Delphi process. RESULTS Participants from 49 countries submitted 336 valid responses. Hormonal therapy for 3 to 6 months was suggested before surgical sperm retrieval (SSR) by 29.6% and 23.6% of participants for normogonadotropic hypogonadism and hypergonadotropic hypogonadism respectively. The SSR rate was reported as 50.0% by 26.0% to 50.0% of participants. Interestingly, 46.0% reported successful SSR in <10% of men with Klinefelter syndrome and 41.3% routinely recommended preimplantation genetic testing. Varicocele repair prior to SSR is recommended by 57.7%. Half of the respondents (57.4%) reported using ultrasound to identify the most vascularized areas in the testis for SSR. One-third proceed directly to microdissection testicular sperm extraction (mTESE) in every case of NOA while others use a staged approach. After a failed conventional TESE, 23.8% wait for 3 months, while 33.1% wait for 6 months before proceeding to mTESE. The cut-off of follicle-stimulating hormone for positive SSR was reported to be 12-19 IU/mL by 22.5% of participants and 20-40 IU/mL by 27.8%, while 31.8% reported no upper limit. CONCLUSIONS This is the largest survey to date on the real-world medical and surgical management of NOA by reproductive experts. It demonstrates a diverse practice pattern and highlights the need for evidence-based international consensus guidelines.
Collapse
Affiliation(s)
- Amarnath Rambhatla
- Department of Urology, Henry Ford Health System, Vattikuti Urology Institute, Detroit, MI, USA
| | - Rupin Shah
- Division of Andrology, Department of Urology, Lilavati Hospital and Research Centre, Mumbai, India
| | - Imad Ziouziou
- Department of Urology, College of Medicine and Pharmacy, Ibn Zohr University, Agadir, Morocco
| | - Priyank Kothari
- Department of Urology, Topiwala National Medical College, B.Y.L Nair Charitable Hospital, Mumbai, India
| | - Gianmaria Salvio
- Department of Endocrinology, Polytechnic University of Marche, Ancona, Italy
| | - Murat Gul
- Department of Urology, Selçuk University School of Medicine, Konya, Turkey
| | - Taha Hamoda
- Department of Urology, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Urology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Parviz Kavoussi
- Department of Reproductive Urology, Austin Fertility & Reproductive Medicine/Westlake IVF, Austin, TX, USA
| | - Widi Atmoko
- Department of Urology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Tuncay Toprak
- Department of Urology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ponco Birowo
- Department of Urology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Edmund Ko
- Department of Urology, Loma Linda University Health, Loma Linda, CA, USA
| | - Mohamed Arafa
- Department of Urology, Hamad Medical Corporation, Doha, Qatar
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Urology, Weill Cornell Medical-Qatar, Doha, Qatar
| | - Ramy Abou Ghayda
- Urology Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, USA
| | | | - Giorgio Ivan Russo
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | - Eric Chung
- Department of Urology, Princess Alexandra Hospital, University of Queensland, Brisbane, Australia
| | - Ahmed M. Harraz
- Department of Urology, Mansoura University Urology and Nephrology Center, Mansoura, Egypt
- Department of Surgery, Urology Unit, Farwaniya Hospital, Farwaniya, Kuwait
- Department of Urology, Sabah Al Ahmad Urology Center, Kuwait City, Kuwait
| | - Marlon Martinez
- Section of Urology, Department of Surgery, University of Santo Tomas Hospital, Manila, Philippines
| | | | - Nicholas Tadros
- Division of Urology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Ramadan Saleh
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Missy Savira
- Department of Urology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Giovanni M. Colpi
- Andrology and IVF Center, Next Fertility Procrea, Lugano, Switzerland
| | - Wael Zohdy
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Edoardo Pescatori
- Andrology and Reproductive Medicine Unit, Next Fertility GynePro, Bologna, Italy
| | - Hyun Jun Park
- Department of Urology, Pusan National University School of Medicine, Busan, Korea
- Medical Research Institute of Pusan National University Hospital, Busan, Korea
| | - Shinichiro Fukuhara
- Department of Urology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Akira Tsujimura
- Department of Urology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Cesar Rojas-Cruz
- Department of Urology, University Hospital of Rostock, Rostock, Germany
| | - Angelo Marino
- Reproductive Medicine Unit, ANDROS Day Surgery Clinic, Palermo, Italy
| | - Siu King Mak
- Department of Surgery, Union Hospital Reproductive Medicine Centre (Tsim Sha Tsui), Kowloon, China
| | - Edouard Amar
- Department of Urology, American Hospital of Paris, Paris, France
| | - Wael Ibrahim
- Department of Obstetrics Gynecology and Reproductive Medicine, Fertility Care Center in Cairo, Cairo, Egypt
| | - Puneet Sindhwani
- Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Naif Alhathal
- Department of Urology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Gian Maria Busetto
- Department of Urology and Organ Transplantation, University of Foggia, Foggia, Italy
| | - Manaf Al Hashimi
- Department of Urology, Burjeel Hospital, Abu Dhabi, UAE
- Department of Urology, Khalifa University College of Medicine and Health Science, Abu Dhabi, UAE
| | - Ahmed El-Sakka
- Department of Urology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Asci Ramazan
- 45Department of Urology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Fotios Dimitriadis
- 1st Urology Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Massimiliano Timpano
- Department of Urology, Molinette Hospital, A.O.U. Città della Salute e della Scienza, University of Turin, Torino, Italy
| | - Davor Jezek
- Department for Transfusion Medicine and Transplantation Biology, Reproductive Tissue Bank, University Hospital Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Baris Altay
- Department of Urology, Ege University Medical School, Bornova, Turkey
| | - Daniel Suslik Zylbersztejn
- Department of Surgery, Discipline of Urology, Fleury Group and Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Michael YC Wong
- Department of Andrology, International Urology, Fertility and Gynecology Centre, Mount Elizabeth Hospital, Singapore
| | - Du Geon Moon
- Department of Urology, Korea University Guro Hospital, Seoul, Korea
| | - Christine Wyns
- Department of Gynaecology-Andrology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Safar Gamidov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Ministry of Health of Russia, Moscow, Russia
| | - Hamed Akhavizadegan
- Department of Urology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Kaan Aydos
- Department of Urology, Ankara University, Ankara, Turkey
| | - Nguyen Quang
- Center for Andrology and Sexual Medicine, Viet Duc University Hospital, Hanoi, Vietnam
- Department of Urology, Andrology and Sexual Medicine, University of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
| | - Shedeed Ashour
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | | - Sava Micic
- Department of Andrology, Uromedica Polyclinic, Belgrade, Serbia
| | - Saleh Binsaleh
- Division of Urology, Deparment of Surgery, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Alayman Hussein
- Department of Urology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Haitham Elbardisi
- Department of Urology, Hamad Medical Corporation, Doha, Qatar
- Department of Urology, Weill Cornell Medical-Qatar, Doha, Qatar
| | - Taymour Mostafa
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | | - Islam Fathy Soliman Abdelrahman
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Andrology, Armed Forces College of Medicine, Cairo, Egypt
| | - Osvaldo Rajmil
- Department of Andrology, Fundació Puigvert, Barcelona, Spain
| | - Arif Kalkanli
- Department of Urology, Taksim Education and Research Hospital, Istanbul, Turkey
| | | | - Kadir Bocu
- Urology Department, Niğde Omer Halis Demir University, Faculty of Medicine, Sirnak, Turkey
| | | | - Gökhan Çeker
- Department of Urology, Başakşehir Çam and Sakura City Hospital, Istanbul, Turkey
| | - Ege Can Serefoglu
- Department of Urology, Biruni University School of Medicine, Istanbul, Turkey
| | - Fahmi Bahar
- Andrology Section, Siloam Sriwijaya Hospital, Palembang, Indonesia
| | - Nazim Gherabi
- Department of Medicine, University of Algiers 1, Algiers, Algeria
| | - Shinnosuke Kuroda
- Department of Urology, Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | - Ahmet Gudeloglu
- Department of Urology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Erman Ceyhan
- Department of Urology, Faculty of Medicine, Baskent University, Ankara, Turkey
| | - Mohamed Saeed Mohamed Hasan
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Muhammad Ujudud Musa
- Urology Unit, Department of Surgery, Federal Medical Center, Katsina State, Nigeria
| | - Ahmad Motawi
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Chak-Lam Cho
- Department of Surgery, S. H. Ho Urology Centre, The Chinese University of Hong Kong, Hong Kong
| | - Hisanori Taniguchi
- Department of Urology and Andrology, Kansai Medical University, Osaka, Japan
| | | | | | - Shingai Mutambirwa
- Department of Urology, Dr. George Mukhari Academic Hospital, Sefako Makgatho Health Science University, Medunsa, South Africa
| | - Nur Dokuzeylul Gungor
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and IVF Unit, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Marion Bendayan
- Department of Reproductive Biology, Fertility Preservation, Andrology, CECOS, Poissy Hospital, Poissy, France
- Department of Biology, Reproduction, Epigenetics, Environment and Development, Paris Saclay University, UVSQ, INRAE, BREED, Jouy-en-Josas, France
| | - Carlo Giulioni
- Department of Urology, Polytechnic University of Marche, Ancona, Italy
| | - Aykut Baser
- Department of Urology, Faculty of Medicine, Bandırma Onyedi Eylül University, Balıkesir, Turkey
| | - Marco Falcone
- Department of Urology, Molinette Hospital, A.O.U. Città della Salute e della Scienza, University of Turin, Torino, Italy
| | - Luca Boeri
- Department of Urology, IRCCS Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Gideon Blecher
- Department of Surgery, School of Clinical Sciences, Monash University, Melbourne, Australia
- Department of Urology, The Alfred Hospital, Melbourne, Australia
| | - Alireza Kheradmand
- Department of Urology, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Ricky Adriansjah
- Department of Urology, Hasan Sadikin General Hospital, Faculty of Medicine of Padjadjaran University, Bandung, Indonesia
| | - Nima Narimani
- Department of Urology, School of Medicine, Hasheminejad Kidney Center, Iran University of Medical Science, Tehran, Iran
| | | | - Tuan Thanh Nguyen
- Department of Urology, University of California, Irvine, CA, USA
- Department of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Department of Urology, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - Andrian Japari
- Department of Urology, Fertility Clinic, Telogorejo Hospital, Central Java, Indonesia
| | - Parisa Dolati
- Department of Animal Science, Faculty of Agriculture, University of Shiraz, Shiraz, Iran
| | - Keerti Singh
- Department of Preclinical and Health Sciences, Faculty of Medical Sciences, The University of West Indies, Bridgetown, Barbados
- Windsor Medical Centre, Bridgetown, Barbados
| | - Cevahir Ozer
- Department of Urology, Faculty of Medicine, Baskent University, Ankara, Turkey
| | - Selcuk Sarikaya
- Department of Urology, Gulhane Research and Training Hospital, University of Health Sciences, Ankara, Turkey
| | - Nadia Sheibak
- Department of Anatomical Sciences, Reproductive Sciences and Technology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Ndagijimana Jean Bosco
- Department of Dermatology, Venereology & Andrology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | | | - Sang Thanh Le
- Department of Urology, Faculty of Medicine, Minia University, Minia, Egypt
- Department of Urology, Fertility Clinic, Telogorejo Hospital, Central Java, Indonesia
| | - Ioannis Sokolakis
- Department of Urology, Martha-Maria Hospital Nuremberg, Nuremberg, Germany
| | - Darren Katz
- Men’s Health Melbourne, Victoria, Australia
- Department of Surgery, Western Precinct, University of Melbourne, Victoria, Australia
- Department of Urology, Western Health, Victoria, Australia
| | - Ryan Smith
- Department of Urology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Manh Nguyen Truong
- Department of Animal Science, Faculty of Agriculture, University of Shiraz, Shiraz, Iran
- Fertility Centre, Hanh Phuc International Hospital, Binh Duong, Vietnam
| | - Tan V. Le
- Department of Andrology, Binh Dan Hospital, Ho Chi Minh City, Vietnam
- Department of Urology and Andrology, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | - Zhongwei Huang
- Department of Obstetrics and Gynaecology, National University Health Systems, Singapore
| | - Muslim Dogan Deger
- Department of Urology, Edirne Sultan 1st Murat State Hospital, Edirne, Turkey
| | - Umut Arslan
- Department of Urology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Gokhan Calik
- Department of Urology, Istanbul Medipol University, Istanbul, Turkey
| | - Giorgio Franco
- Department of Urology, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Ayman Rashed
- 123Department of Urology, Faculty of Medicine, 6th of October University, Giza, Egypt
| | - Oguzhan Kahraman
- Department of Urology, Faculty of Medicine, Baskent University, Ankara, Turkey
| | | | - Rosadi Putra
- Department of Urology, RSUD Ciawi Regional General Hospital, West Java, Indonesia
| | - Giancarlo Balercia
- Department of Endocrinology, Polytechnic University of Marche, Ancona, Italy
| | - Kareim Khalafalla
- Department of Urology, Princess Alexandra Hospital, University of Queensland, Brisbane, Australia
- Department of Urology, University of Illinois, Chicago, IL, USA
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Anh Đặng Tuấn
- Tam Anh IVF Center, Tam Anh General Hospital, Hanoi, Vietnam
| | - Amr El Meliegy
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Birute Zilaitiene
- Institute of Endocrinology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | - Filippo Giacone
- HERA Center, Unit of Reproductive Medicine, Sant'Agata Li Battiati, Catania, Italy
| | - Aldo E. Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | - Sunil Jindal
- Department of Andrology & Reproductive Medicine, Jindal Hospital & Fertility Center, Meerut, India
| | - Bac Nguyen Hoai
- Department of Andrology and Sexual Medicine, Hanoi Medical University Hospital, Hanoi, Vietnam
| | - Ravi Banthia
- Department of Urology, Western General Hospital, Edinburgh, UK
| | - Marcelo Rodriguez Peña
- Institute of Gynecology and Fertility (IFER), University of Buenos Aires, Buenos Aires, Argentina
| | - Dharani Moorthy
- IVF Department, Swarupa Fertility & IVF Centre, Vijayawada, India
| | - Aram Adamyan
- Department of Urology, Astghik Medical Center, Yerevan, Armenia
| | - Deniz Kulaksiz
- Department of Obstetrics and Gynecology, Kanuni Training and Research Hospital, University of Health Sciences, Trabzon, Turkey
| | | | - Nikolaos Sofikitis
- Department of Urology, Ioannina University School of Medicine, Ioannina, Greece
| | - Ciro Salzano
- PO San Giovanni Bosco, ASL Napoli 1 Centro, Napoli, Italy
| | | | - Surendra Reddy Banka
- Department of Andrology, Androcare Institute of Andrology and Men's Health, Hyderabad, India
| | - Tiago Cesar Mierzwa
- Department of Urology, Centro Universitario em Saude do ABC, Santo André, Brazil
| | - Tahsin Turunç
- Urology Clinic, Iskenderun Gelisim Hospital, Iskenderun, Turkey
| | - Divyanu Jain
- Department of Obstetrics and Gynecology, Jaipur Golden Hospital, New Delhi, India
| | - Armen Avoyan
- Department of Obstetrics and Gynecology, Kanuni Training and Research Hospital, University of Health Sciences, Trabzon, Turkey
| | - Pietro Salacone
- Andrology and Pathophysiology of Reproduction Unit, Santa Maria Goretti Hospital, Latina, Italy
| | - Ateş Kadıoğlu
- Section of Andrology, Department of Urology, Istanbul University Faculty of Medicine, Istanbul, Turkey
| | - Chirag Gupta
- Department of Urology, Jaipur National University, Jaipur, India
| | - Haocheng Lin
- Department of Urology, Peking University Third Hospital, Peking University, Beijing, China
| | - Iman Shamohammadi
- Department of Urology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasser Mogharabian
- Sexual Health and Fertility Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | | | - Yavuz Onur Danacıoğlu
- Department of Urology, University of Health Science, Istanbul Bakırköy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Salima Daoud
- Laboratory of Histo-Embryology and Reproductive Biology, Faculty of Medicine of Sfax, University of Sfax, Sfax, Tunisia
| | - Vineet Malhotra
- Department of Urology and Andrology, VNA Hospital, New Delhi, India
| | - Abdulmalik Almardawi
- Department of Urology, Prince Sultan Millitary Medical City, Riyadh, Saudi Arabia
| | - Osama Mohamed Selim
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamad Moussa
- Department of Urology, Lebanese University, Beirut, Lebanon
- Department of Urology, Al Zahraa Hospital, UMC, Beirut, Lebanon
| | - Saeid Haghdani
- Andrology Research Center, Yazd Reproductive Science Institute, Isfahan Fertility and Infertility Center, Isfahan, Iran
| | - Mesut Berkan Duran
- Department of Urology, Pamukkale University School of Medicine, Denizli, Turkey
| | - Yannic Kunz
- Department of Urology, University Hospital Innsbruck, Innsbruck, Austria
| | - Mirko Preto
- Department of Urology, Molinette Hospital, A.O.U. Città della Salute e della Scienza, University of Turin, Torino, Italy
| | - Elena Eugeni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Department of Medicine and Medical Specialties, Division of Medical Andrology and Endocrinology of Reproduction, University of Terni, Terni, Italy
| | - Thang Nguyen
- Department of Obstetrics and Gynecology, Kanuni Training and Research Hospital, University of Health Sciences, Trabzon, Turkey
| | - Ahmed Rashad Elshahid
- 123Department of Urology, Faculty of Medicine, 6th of October University, Giza, Egypt
| | | | - Dyandra Parikesit
- Department of Urology, Faculty of Medicine, Universitas Indonesia Hospital, Depok, Indonesia
| | - Essam Nada
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | | | - Florence Boitrelle
- Department of Reproductive Biology, Fertility Preservation, Andrology, CECOS, Poissy Hospital, Poissy, France
- Department of Biology, Reproduction, Epigenetics, Environment and Development, Paris Saclay University, UVSQ, INRAE, BREED, Jouy-en-Josas, France
| | | | - Mounir Jamali
- Department of Urology, Military Teaching Hospital, Rabat, Morocco
| | - Raju Nair
- Department of Reproductive Medicine, Mitera Hospital, Kottayam, India
| | | | - Franco Gadda
- Department of Urology, IRCCS Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Charalampos Thomas
- Urology and Neuro-Urology Unit, National Rehabilitation Center, Athens, Greece
| | | | - Umit Gul
- Private EPC Hospital, Adana, Turkey
| | - Serena Maruccia
- Department of Urology, ASST Santi Paolo e Carlo, San Paolo Hospital, Milano, Italy
| | - Ajay Kanbur
- Department of Andrology, Kanbur Clinic, Thane, India
- Department of Urosurgery, Jupiter Hospital, Thane, India
| | | | | | - Raghavender Kosgi
- Department of Andrology and Men’s Health, Apollo Hospitals, Hyderabad, India
| | - Fatih Gokalp
- Department of Urology, Faculty of Medicine, Hatay Mustafa Kemal University, Antakya, Turkey
| | | | - Gustavo Marquesine Paul
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Hesamoddin Sajadi
- Department of Urology, Bombay Hospital and Medical Research Center, Mumbai, India
| | - Deepak Gupte
- Department of Urology, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | - Rafael F. Ambar
- Department of Urology, Centro Universitario em Saude do ABC, Santo André, Brazil
| | | | - Karun Singla
- Department of Urology, Dr. Dradjat Hospital, Serang, Indonesia
| | | | - Shannon Hee Kyung Kim
- Department of Urology, Macquarie University Faculty of Medicine and Health Sciences, Sydney, Australia
| | | | - Koichi Nagao
- Department of Urology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Sakti Ronggowardhana Brodjonegoro
- Division of Urology, Department of Surgery, Prof. Dr. Sardjito Hospital, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Andri Rezano
- Andrology Study Program, Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia
| | | | - Rossella Mazzilli
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Hasan M. A. Farsi
- Department of Urology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hung Nguyen Ba
- Andrology Unit, ART Center, Vinmec Times City International Hospital, Hanoi, Vietnam
| | - Hamed Alali
- Department of Urology, Macquarie University Faculty of Medicine and Health Sciences, Sydney, Australia
| | | | - Tran Quang Tien Long
- Department of Obstetrics and Gynecology, Hanoi Obstetrics and Gynecology Hospital, Hanoi, Vietnam
| | - Sami Alsaid
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hoang Bao Ngoc Cuong
- Department of Surgery, Hai Phong University of Medicine and Pharmacy, Hai Phong, Vietnam
| | - Knigavko Oleksandr
- Department of Urology, Nephrology and Andrology Kharkiv National Medical University, Kharkiv, Ukraine
| | - Akhmad Mustafa
- Department of Urology, Hasan Sadikin General Hospital, Faculty of Medicine of Padjadjaran University, Bandung, Indonesia
| | - Herik Acosta
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | | | - Bahadır Şahin
- Department of Urology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Eko Arianto
- Department of Urology, Prof R.D. Kandou Hospital, Manado, Indonesia
| | - Colin Teo
- Department of Urology, Gleneagles Hospital, Singapore
| | | | - Rinaldo Indra Rachman
- Department of Urology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Mustafa Gurkan Yenice
- Department of Urology, University of Health Science, Istanbul Bakırköy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | | | - Shivam Priyadarshi
- Department of Urology, Sawai Man Singh Medical College and Hospital, Jaipur, Rajasthan, India
| | - Marko Tanic
- Department of Urology, General Hospital, Cuprija, Serbia
| | - Noor Kareem Alfatlaw
- Fertility Center of Al-Najaf, Al-Sadr Medical City, Babylon Health Directorate, Iraqi Ministry of Health, Baghdad, Iraq
| | - Fikri Rizaldi
- Andrology Study Program, Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ranjit B. Vishwakarma
- Division of Andrology, Department of Urology, Lilavati Hospital and Research Centre, Mumbai, India
| | - George Kanakis
- Department of Endocrinology, Diabetes and Metabolism, Athens Naval & VA Hospital, Athens, Greece
| | | | - Joe Lee
- Department of Urology, National University Hospital, Singapore
| | - Raisa Galstyan
- Department of Urology, Yerevan State Medical University, Yerevan, Armenia
| | - Hakan Keskin
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Janan Wurzacher
- Department of Urology, University Hospital Innsbruck, Innsbruck, Austria
| | - Doddy Hami Seno
- Division of Urology, Department of Surgery, Persahabatan General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Bambang S Noegroho
- Department of Urology, Hasan Sadikin General Hospital, Faculty of Medicine of Padjadjaran University, Bandung, Indonesia
| | - Ria Margiana
- Department of Urology, Prof R.D. Kandou Hospital, Manado, Indonesia
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Master’s Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Urology Unit, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Qaisar Javed
- Department of Urology, Al-Ahlia Hospital, Abu Dhabi, UAE
| | | | | | - Ana Puigvert
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and IVF Unit, School of Medicine, Bahcesehir University, Istanbul, Turkey
- Institute of Andrology and Sexual Medicine (IANDROMS), Barcelona, Spain
| | - Coşkun Kaya
- Department of Urology, Health Science University Eskisehir City HPRH, Eskisehir, Turkey
| | | | - Chadi Yazbeck
- Department of Obstetrics Gynecology and Reproductive Medicine, Reprogynes Medical Institute, Paris, France
| | - Azwar Amir
- Department of Urology, Dr Wahidin Sudirohusodo Hospital, Makassar, Indonesia
| | - Edson Borges
- IVF Department, Fertility Assisted Fertilization Center, São Paulo, Brazil
| | - Marina Bellavia
- Andrology and IVF Center, Next Fertility Procrea, Lugano, Switzerland
| | - Isaac Ardianson Deswanto
- Department of Urology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Vinod KV
- Department of Urology, Cure & SK Hospital, Trivandrum, India
| | | | - Dang Hoang Minh
- Department of Urology, Andrology and Sexual Medicine, University of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
| | | | - Fulvio Colombo
- Andrology and Reproductive Medicine Unit, Next Fertility GynePro, Bologna, Italy
| | - Armand Zini
- Department of Surgery, McGill University, Montreal, QC, Canada
| | - Niket Patel
- Akanksha Hospital and Research Institute, Anand, Gujarat, India
| | - Selahittin Çayan
- Department of Urology, University of Mersin School of Medicine, Mersin, Turkey
| | - Ula Al-kawaz
- High Institute for Infertility Diagnosis and Assisted Reproductive Technologies, Al-Nahrain University, Baghdad, Iraq
| | - Maged Ragab
- Department of Andrology, Tanta University, Tanta, Egypt
| | | | | | - Ozan Efesoy
- Department of Andrology, Tanta University, Tanta, Egypt
| | - Ivan Hoffmann
- Department of Reproductive Medicine and Andrology, University Clinic Halle (Saale), Halle, Germany
- Reproductive Center Dr. Hoffmann, Berlin, Germany
| | - Thiago Afonso Teixeira
- Division of Urology, University Hospital, Federal University of Amapa, Macapá, Brazil
- Men’s Health Study Group, Institute for Advanced Studies, University of São Paulo, São Paulo, Brazil
- Androscience-Science and Innovation Center and High Complexity Clinical and Research Andrology Laboratory, São Paulo, Brazil
| | - Barış Saylam
- Department of Urology, University of Mersin School of Medicine, Mersin, Turkey
| | | | | | | |
Collapse
|
2
|
Khaydukova IV, Ivannikova VM, Zhidkov DA, Belikov NV, Peshkova MA, Timashev PS, Tsiganov DI, Pushkarev AV. Current State and Challenges of Tissue and Organ Cryopreservation in Biobanking. Int J Mol Sci 2024; 25:11124. [PMID: 39456905 PMCID: PMC11508709 DOI: 10.3390/ijms252011124] [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: 08/16/2024] [Revised: 09/26/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Recent years have witnessed significant advancements in the cryopreservation of various tissues and cells, yet several challenges persist. This review evaluates the current state of cryopreservation, focusing on contemporary methods, notable achievements, and ongoing difficulties. Techniques such as slow freezing and vitrification have enabled the successful preservation of diverse biological materials, including embryos and ovarian tissue, marking substantial progress in reproductive medicine and regenerative therapies. These achievements highlight improved post-thaw survival and functionality of cryopreserved samples. However, there are remaining challenges such as ice crystal formation, which can lead to cell damage, and the cryopreservation of larger, more complex tissues and organs. This review also explores the role of cryoprotectants and the importance of optimizing both cooling and warming rates to enhance preservation outcomes. Future research priorities include developing new cryoprotective agents, elucidating the mechanisms of cryoinjury, and refining protocols for preserving complex tissues and organs. This comprehensive overview underscores the transformative potential of cryopreservation in biomedicine, while emphasizing the necessity for ongoing innovation to address existing challenges.
Collapse
Affiliation(s)
- Irina V. Khaydukova
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
| | - Valeria M. Ivannikova
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
| | - Dmitry A. Zhidkov
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
| | - Nikita V. Belikov
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
| | - Maria A. Peshkova
- Institute for Regenerative Medicine, Sechenov University, 119048 Moscow, Russia
| | - Peter S. Timashev
- Institute for Regenerative Medicine, Sechenov University, 119048 Moscow, Russia
| | - Dmitry I. Tsiganov
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
- Russian Medical Academy of Continuous Professional Education, 125993 Moscow, Russia
| | - Aleksandr V. Pushkarev
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
- Russian Medical Academy of Continuous Professional Education, 125993 Moscow, Russia
| |
Collapse
|
3
|
Bashiri Z, Hosseini SJ, Salem M, Koruji M. In vivo and in vitro sperm production: an overview of the challenges and advances in male fertility restoration. Clin Exp Reprod Med 2024; 51:171-180. [PMID: 38525520 PMCID: PMC11372308 DOI: 10.5653/cerm.2023.06569] [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: 10/03/2023] [Accepted: 12/14/2023] [Indexed: 03/26/2024] Open
Abstract
Male infertility can be caused by genetic anomalies, endocrine disorders, inflammation, and exposure to toxic chemicals or gonadotoxic treatments. Therefore, several recent studies have concentrated on the preservation and restoration of fertility to enhance the quality of life for affected individuals. It is currently recommended to biobank the tissue extracted from testicular biopsies to provide a later source of spermatogonial stem cells (SSCs). Another successful approach has been the in vitro production of haploid male germ cells. The capacity of SSCs to transform into sperm, as in testicular tissue transplantation, SSC therapy, and in vitro or ex vivo spermatogenesis, makes them ideal candidates for in vivo fertility restoration. The transplantation of SSCs or testicular tissue to regenerate spermatogenesis and create embryos has been achieved in nonhuman mammal species. Although the outcomes of human trials have yet to be released, this method may soon be approved for clinical use in humans. Furthermore, regenerative medicine techniques that develop tissue or cells on organic or synthetic scaffolds enriched with bioactive molecules have also gained traction. All of these methods are now in different stages of experimentation and clinical trials. However, thanks to rigorous studies on the safety and effectiveness of SSC-based reproductive treatments, some of these techniques may be clinically available in upcoming decades.
Collapse
Affiliation(s)
- Zahra Bashiri
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Omid Fertility and Infertility Clinic, Hamedan, Iran
| | - Seyed Jamal Hosseini
- Biomedical Engineering Department, Amirkabir University of Technology, Tehran, Iran
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Salem
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Koruji
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Sung ZY, Liao YQ, Hou JH, Lai HH, Weng SM, Jao HW, Lu BJ, Chen CH. Advancements in fertility preservation strategies for pediatric male cancer patients: a review of cryopreservation and transplantation of immature testicular tissue. Reprod Biol Endocrinol 2024; 22:47. [PMID: 38637872 PMCID: PMC11025181 DOI: 10.1186/s12958-024-01219-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 04/06/2024] [Indexed: 04/20/2024] Open
Abstract
Recently, there has been increasing emphasis on the gonadotoxic effects of cancer therapy in prepubertal boys. As advances in oncology treatments continue to enhance survival rates for prepubertal boys, the need for preserving their functional testicular tissue for future reproduction becomes increasingly vital. Therefore, we explore cutting-edge strategies in fertility preservation, focusing on the cryopreservation and transplantation of immature testicular tissue as a promising avenue. The evolution of cryopreservation techniques, from controlled slow freezing to more recent advancements in vitrification, with an assessment of their strengths and limitations was exhibited. Detailed analysis of cryoprotectants, exposure times, and protocols underscores their impact on immature testicular tissue viability. In transplantation strategy, studies have revealed that the scrotal site may be the preferred location for immature testicular tissue grafting in both autotransplantation and xenotransplantation scenarios. Moreover, the use of biomaterial scaffolds during graft transplantation has shown promise in enhancing graft survival and stimulating spermatogenesis in immature testicular tissue over time. This comprehensive review provides a holistic approach to optimize the preservation strategy of human immature testicular tissue in the future.
Collapse
Affiliation(s)
- Zih-Yi Sung
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Yong-Qi Liao
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Jung-Hsiu Hou
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan, ROC
| | - Hong-Hsien Lai
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Sung-Ming Weng
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Hai-Wei Jao
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan, ROC
| | - Buo-Jia Lu
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan, ROC
| | - Chi-Huang Chen
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan, ROC.
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC.
| |
Collapse
|
5
|
Albamonte MI, Vitullo AD. Preservation of fertility in female and male prepubertal patients diagnosed with cancer. J Assist Reprod Genet 2023; 40:2755-2767. [PMID: 37770817 PMCID: PMC10656407 DOI: 10.1007/s10815-023-02945-2] [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: 07/04/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023] Open
Abstract
Over the past two decades, the importance of fertility preservation has grown not only in the realm of medical and clinical patient care, but also in the field of basic and applied research in human reproduction. With advancements in cancer treatments resulting in higher rates of patient survival, it is crucial to consider the quality of life post-cure. Therefore, fertility preservation must be taken into account prior to antitumor treatments, as it can significantly impact a patient's future fertility. For postpubertal patients, gamete cryopreservation is the most commonly employed preservation strategy. However, for prepubertal patients, the situation is more intricate. Presently, ovarian tissue cryopreservation is the standard practice for prepubertal girls, but further scientific evidence is required in several aspects. Testicular tissue cryopreservation, on the other hand, is still experimental for prepubertal boys. The primary aim of this review is to address the strategies available for possible fertility preservation in prepubertal girls and boys, such as ovarian cryopreservation/transplantation, in vitro follicle culture and meiotic maturation, artificial ovary, transplantation of cryopreserved spermatogonia, and cryopreservation/grafting of immature testicular tissue and testicular organoids.
Collapse
Affiliation(s)
- María Itatí Albamonte
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, C1405BCK, Buenos Aires, Argentina
| | - Alfredo D Vitullo
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, C1405BCK, Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
6
|
Anvari A, Movahedin M, Hamzeh M. Optimizing Immature Testicular Tissue and Cell Transplantation Results: Comparing Transplantation Sites and Scaffolds. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2023; 18:12-19. [PMID: 38041454 PMCID: PMC10692742 DOI: 10.22074/ijfs.2023.559999.1350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 05/06/2023] [Accepted: 06/27/2023] [Indexed: 12/03/2023]
Abstract
For patients who had testicular tissue cryopreserved before receiving gonadotoxic therapies, transplantation of testicular tissues and cells has been recommended as a potential therapeutic option. There are no studies that indicate the generation of sperm after human immature testicular tissue (ITT) or spermatogonial stem cells (SSCs) transplantation. The use of releasing scaffolds and localized drug delivery systems as well as the optimizing transplantation site can play an effective role in increasing the efficiency and improving the quality of testicular tissue and cell transplantation in animal models. Current research is focused on optimizing ITT and cell transplantation, the use of releasing scaffolds, and the selection of the right transplantation site that might restore sperm production or male infertility treatment. By searching the PubMed and Google Scholar databases, original and review papers were collected. Search terms were relevant for SSCs and tissue transplantation. In this review, we'll focus on the potential advantages of using scaffolds and choosing the right transplantation site to improve transplantation outcomes.
Collapse
Affiliation(s)
- Alireza Anvari
- Department of Anatomical Sciences, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Mansoureh Movahedin
- Department of Anatomical Sciences, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran.
| | - Maedeh Hamzeh
- Department of Anatomical Sciences, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
7
|
Gomes FDR, Ñaupas LVS, Palomino GJQ, Celiz RHY, Sá NAR, Novaes MAS, Ferreira ACA, Brito DCC, Freitas VJF, Costa BN, Lucci CM, Fernandes CCL, Rondina D, Figueiredo JR, Tetaping GM, Rodrigues APR. Definition of protocols for cryopreservation and three-dimensional in vitro culture of prepubertal goat testicular tissue after histomorphological, ultrastructural, and functional analysis. Theriogenology 2023; 211:151-160. [PMID: 37639997 DOI: 10.1016/j.theriogenology.2023.08.015] [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: 04/20/2023] [Revised: 07/27/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023]
Abstract
This study aims to define the best method (slow freezing or vitrification) and fragment size (1, 5, or 9 mm³) for prepubertal goat testis cryopreservation, as well as to evaluate testicular morphological integrity after cryopreservation and in vitro culture (IVC). Initially (experiment I), 1, 5, or 9 mm³ testis fragments were cryopreserved by slow freezing using a Mr. Frosty container with 20% Dimethylsulfoxide (DMSO) or vitrified using the Ovarian Tissue Cryosystem (OTC) device, (Equilibration solution - ES: 10% DMSO and 10% ethylene glycol - EG; Vitrification solution - VS: 20% DMSO and 20% EG) and then subjected to morphological analysis, type I and III collagen quantification and gene expression (Oct4, C-kit, Bax, and Bcl-2). Subsequently, (experiment II), fresh or cryopreserved by slow freezing testis fragments were cultured in vitro and submitted to morphological analysis by scanning electron microscopy. The data from the experiment I revealed fewer morphological alterations in 1 and 5 mm³ fragments after vitrification and slow freezing, respectively. The percentage of type I collagen fibers in 5 and 9 mm³ frozen was higher than in fresh or vitrified fragments. For type III collagen, fresh or frozen fragments of 1 and 5 mm3 showed a higher percentage than fragments of 9 mm3. Gene expression for Oct4 and C-kit after slow freezing or vitrification in the 5 mm3 fragments was lower than that observed in the fresh fragments. The Bax:Bcl-2 ratio in the 1 and 9 mm³ fragments was lower than in the 5 mm³ fragments for fresh fragments or after freezing. In experiment II, fragments cultured in vitro, previously frozen or not, showed more morphological alterations than fresh or frozen fragments. We concluded that slow freezing of 5 mm³ fragments was the best protocol for cryopreserving prepubertal goat testis and although the results of IVC are encouraging, it still needs improvement to restore testicular function after cryopreservation.
Collapse
Affiliation(s)
- F D R Gomes
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - L V S Ñaupas
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - G J Q Palomino
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - R H Y Celiz
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - N A R Sá
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - M A S Novaes
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - A C A Ferreira
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - D C C Brito
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - V J F Freitas
- Laboratory of Physiology and Control of Reproduction (LFCR), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - B N Costa
- Laboratory of Animal Reproduction, Department of Physiological Sciences, Institute of Biological Sciences, Darcy Ribeiro University Campus, Brasília, DF, Brazil
| | - C M Lucci
- Laboratory of Animal Reproduction, Department of Physiological Sciences, Institute of Biological Sciences, Darcy Ribeiro University Campus, Brasília, DF, Brazil
| | - C C L Fernandes
- College of Health Sciences, University of Fortaleza, Fortaleza, CE, Brazil
| | - D Rondina
- Laboratory of Nutrition and Production of Ruminants (LANUPRUMI), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - J R Figueiredo
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - G M Tetaping
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - A P R Rodrigues
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
8
|
Goszczynski DE, Navarro M, Mutto AA, Ross PJ. Review: Embryonic stem cells as tools for in vitro gamete production in livestock. Animal 2023; 17 Suppl 1:100828. [PMID: 37567652 DOI: 10.1016/j.animal.2023.100828] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/13/2023] [Accepted: 04/20/2023] [Indexed: 08/13/2023] Open
Abstract
The goal of in vitro gametogenesis is to reproduce the events of sperm and oocyte development in the laboratory. Significant advances have been made in the mouse in the last decade, but evolutionary divergence from the murine developmental program has prevented the replication of these advances in large mammals. In recent years, intensive work has been done in humans, non-human primates and livestock to elucidate species-specific differences that regulate germ cell development, due to the number of potential applications. One of the most promising applications is the use of in vitro gametes to optimize the spread of elite genetics in cattle. In this context, embryonic stem cells have been posed as excellent candidates for germ cell platforms. Here, we present the most relevant advances in in vitro gametogenesis of interest to livestock science, including new types of pluripotent stem cells with potential for germline derivation, characterization of the signaling environment in the gonadal niche, and experimental systems used to reproduce different stages of germ cell development in the laboratory.
Collapse
Affiliation(s)
- D E Goszczynski
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo Ugalde"- UNSAM-CONICET, Buenos Aires CP 1650, Argentina
| | - M Navarro
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo Ugalde"- UNSAM-CONICET, Buenos Aires CP 1650, Argentina
| | - A A Mutto
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo Ugalde"- UNSAM-CONICET, Buenos Aires CP 1650, Argentina
| | - P J Ross
- Department of Animal Science, University of California Davis, Davis, CA, USA; STgenetics, Navasota, TX, USA.
| |
Collapse
|
9
|
Silva AR, Pereira AF, Comizzoli P. Biobanking and use of gonadal tissues - a promising strategy for conserving wildlife from the Caatinga biome. Anim Reprod 2023; 19:e20220135. [PMID: 36819484 PMCID: PMC9924771 DOI: 10.1590/1984-3143-ar2022-0135] [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: 01/02/2023] [Accepted: 01/24/2023] [Indexed: 02/19/2023] Open
Abstract
Biological Resource Banks (BRB) or Genetic Resource Banks (GRB) are critical tools for the conservation of animal biodiversity. According to the International Union for Conservation of Nature, more than 38,500 species are threatened with extinction, out of a total of 138,300 surveyed species. These banks are repositories of biological samples and data recovered and preserved for the long term by zoos, universities, research centers and other conservation organizations. In recent years, BRB have increasingly included ovarian and testicular tissues as additional options to rescue and propagate wild species, especially those at risk of extinction. After in vitro culture or grafting, gonadal tissues are potential sources of matured gametes that can be used for Assisted Reproduction Technologies while informing about gametogenesis or mechanisms involved in infertility. It therefore is crucial to properly recover, cryopreserve, and culture these tissues using species-specific protocols. Developing BRBs is currently one of the strategies to preserve species from the Caatinga biome - an exclusively Brazilian biome with a rich wild fauna that suffers from anthropogenic activities. Among wild species from this biome, studies have been primarily conducted in collared peccaries, agoutis, cavies, and armadillos to preserve their ovarian and testicular tissues. Additionally, domestic species such as the domestic cat and donkeys have been proposed as models for wild species that are phylogenetically close. This review addresses the main technical aspects involved in obtaining BRB derived from gonadal tissues in some wild species of the Caatinga biome. It reports recent advances and perspectives to use these biological materials for wildlife conservation.
Collapse
Affiliation(s)
- Alexandre Rodrigues Silva
- Laboratório de Conservação de Germoplasma Animal, Universidade Federal Rural do Semi-Árido, Mossoró, RN, Brasil,Corresponding author:
| | | | - Pierre Comizzoli
- Smithsonian National Zoo and Conservation Biology Institute, Washington, USA
| |
Collapse
|
10
|
Shamhari A‘A, Jefferi NES, Abd Hamid Z, Budin SB, Idris MHM, Taib IS. The Role of Promyelocytic Leukemia Zinc Finger (PLZF) and Glial-Derived Neurotrophic Factor Family Receptor Alpha 1 (GFRα1) in the Cryopreservation of Spermatogonia Stem Cells. Int J Mol Sci 2023; 24:ijms24031945. [PMID: 36768269 PMCID: PMC9915902 DOI: 10.3390/ijms24031945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 01/20/2023] Open
Abstract
The cryopreservation of spermatogonia stem cells (SSCs) has been widely used as an alternative treatment for infertility. However, cryopreservation itself induces cryoinjury due to oxidative and osmotic stress, leading to reduction in the survival rate and functionality of SSCs. Glial-derived neurotrophic factor family receptor alpha 1 (GFRα1) and promyelocytic leukemia zinc finger (PLZF) are expressed during the self-renewal and differentiation of SSCs, making them key tools for identifying the functionality of SSCs. To the best of our knowledge, the involvement of GFRα1 and PLZF in determining the functionality of SSCs after cryopreservation with therapeutic intervention is limited. Therefore, the purpose of this review is to determine the role of GFRα1 and PLZF as biomarkers for evaluating the functionality of SSCs in cryopreservation with therapeutic intervention. Therapeutic intervention, such as the use of antioxidants, and enhancement in cryopreservation protocols, such as cell encapsulation, cryoprotectant agents (CPA), and equilibrium of time and temperature increase the expression of GFRα1 and PLZF, resulting in maintaining the functionality of SSCs. In conclusion, GFRα1 and PLZF have the potential as biomarkers in cryopreservation with therapeutic intervention of SSCs to ensure the functionality of the stem cells.
Collapse
Affiliation(s)
- Asma’ ‘Afifah Shamhari
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Nur Erysha Sabrina Jefferi
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Zariyantey Abd Hamid
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Siti Balkis Budin
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Muhd Hanis Md Idris
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA (UiTM), Puncak Alam Campus, Bandar Puncak Alam 42300, Selangor, Malaysia
| | - Izatus Shima Taib
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
- Correspondence: ; Tel.: +603-928-97608
| |
Collapse
|
11
|
Testicular Tissue Vitrification: a Promising Strategy for Male Fertility Preservation. Reprod Sci 2022; 30:1687-1700. [DOI: 10.1007/s43032-022-01113-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022]
|
12
|
Cryopreservation of testicular tissue from the dog (Canis familiaris) and wild boar (Sus scrofa) by slow freezing and vitrification: Differences in cryoresistance according to cell type. Theriogenology 2022; 190:65-72. [DOI: 10.1016/j.theriogenology.2022.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/20/2022] [Accepted: 07/31/2022] [Indexed: 11/18/2022]
|
13
|
Tracking Immature Testicular Tissue after Vitrification In Vitro and In Vivo for Pre-Pubertal Fertility Preservation: A Translational Transgenic Mouse Model. Int J Mol Sci 2022; 23:ijms23158425. [PMID: 35955560 PMCID: PMC9368802 DOI: 10.3390/ijms23158425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 11/18/2022] Open
Abstract
Pediatric cancer survivors experiencing gonadotoxic chemoradiation therapy may encounter subfertility or permanent infertility. However, previous studies of cryopreservation of immature testicular tissue (ITT) have mainly been limited to in vitro studies. In this study, we aim to evaluate in vitro and in vivo bioluminescence imaging (BLI) for solid surface-vitrified (SSV) ITT grafts until adulthood. The donors and recipients were transgenic and wild-type mice, respectively, with fresh ITT grafts used as the control group. In our study, the frozen ITT grafts remained intact as shown in the BLI, scanning electron microscopy (SEM) and immunohistochemistry (IHC) analyses. Graft survival was analyzed by BLI on days 1, 2, 5, 7, and 31 after transplantation. The signals decreased by quantum yield between days 2 and 5 in both groups, but gradually increased afterwards until day 31, which were significantly stronger than day 1 after transplantation (p = 0.008). The differences between the two groups were constantly insignificant, suggesting that both fresh and SSV ITT can survive, accompanied by spermatogenesis, until adulthood. The ITT in both groups presented similar BLI intensity and intact cells and ultrastructures for spermatogenesis. This translational model demonstrates the great potential of SSV for ITT in pre-pubertal male fertility preservation.
Collapse
|
14
|
Sanou I, van Maaren J, Eliveld J, Lei Q, Meißner A, de Melker AA, Hamer G, van Pelt AMM, Mulder CL. Spermatogonial Stem Cell-Based Therapies: Taking Preclinical Research to the Next Level. Front Endocrinol (Lausanne) 2022; 13:850219. [PMID: 35444616 PMCID: PMC9013905 DOI: 10.3389/fendo.2022.850219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/07/2022] [Indexed: 01/15/2023] Open
Abstract
Fertility preservation via biobanking of testicular tissue retrieved from testicular biopsies is now generally recommended for boys who need to undergo gonadotoxic treatment prior to the onset of puberty, as a source of spermatogonial stem cells (SSCs). SSCs have the potential of forming spermatids and may be used for therapeutic fertility approaches later in life. Although in the past 30 years many milestones have been reached to work towards SSC-based fertility restoration therapies, including transplantation of SSCs, grafting of testicular tissue and various in vitro and ex vivo spermatogenesis approaches, unfortunately, all these fertility therapies are still in a preclinical phase and not yet available for patients who have become infertile because of their treatment during childhood. Therefore, it is now time to take the preclinical research towards SSC-based therapy to the next level to resolve major issues that impede clinical implementation. This review gives an outline of the state of the art of the effectiveness and safety of fertility preservation and SSC-based therapies and addresses the hurdles that need to be taken for optimal progression towards actual clinical implementation of safe and effective SSC-based fertility treatments in the near future.
Collapse
Affiliation(s)
- Iris Sanou
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Jillis van Maaren
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Jitske Eliveld
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Qijing Lei
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Andreas Meißner
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
- Department of Urology, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Annemieke A de Melker
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Geert Hamer
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Ans M M van Pelt
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Callista L Mulder
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
15
|
Kanbar M, Delwiche G, Wyns C. Fertility preservation for prepubertal boys: are we ready for autologous grafting of cryopreserved immature testicular tissue? ANNALES D'ENDOCRINOLOGIE 2022; 83:210-217. [DOI: 10.1016/j.ando.2022.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
16
|
Silva AM, Pereira AG, Bezerra LGP, Jerônimo Moreira SS, Pereira AF, Oliveira MF, Comizzoli P, Silva AR. Cryopreservation of Testicular Tissue from Adult Red-Rumped Agoutis (Dasyprocta leporina Linnaeus, 1758). Animals (Basel) 2022; 12:ani12060738. [PMID: 35327135 PMCID: PMC8944822 DOI: 10.3390/ani12060738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/09/2022] [Accepted: 03/14/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Testicular tissues are composed of many types of germ cells, including early stages which can be grown up to fully formed spermatozoa following grafting or in vitro culture. The systematic banking of testicular tissues at freezing temperatures is useful for future use in assisted reproduction and to improve the reproductive management of rare mammalian species. The present study explored testicular tissue cryopreservation in the red-rumped agouti by slow freezing or vitrification methods, using different combinations of cryoprotectants. Solid-surface vitrification using the combination of dimethyl sulfoxide and ethylene glycol was the most effective protocol to preserve testicular cell morphology and proliferative potential. Abstract This study measured the effects of different freezing techniques and permeating cryoprotectants on the preservation of testicular tissues from adult red-rumped agoutis. Tissue biopsies (3.0 mm3) from five individuals were allocated to different experimental groups: control (non-cryopreserved); slow freezing (SF), solid-surface vitrification (SSV), and conventional vitrification (CV). Each method used dimethyl sulfoxide (DMSO), ethylene glycol (EG), or a DMSO + EG combination. Morphology, viability, mitochondrial activity, and proliferative potential were assessed in fresh and frozen tissue samples. Testicular morphology was better using SSV with a combination of DMSO and EG. Across the different cryopreservation approaches, as well as cryoprotectant combinations, cell viability was comparable. Regarding mitochondrial activity, DMSO + EG/SSV or CV, and DMSO + EG/CV were similar to the EG/SF group, which was the best group that provided values similar to fresh control groups. Adequate preservation of the proliferative potential of spermatogonia, Leydig cells, and Sertoli cells was obtained using SSV with DMSO + EG. Overall, the use of SSV with DMSO + EG was the best protocol for the preservation of testicular tissues from adult red-rumped agoutis.
Collapse
Affiliation(s)
- Andréia M. Silva
- Laboratory of Animal Germplasm Conservation, Department of Animal Sciences, Federal University of Semiarid Region–UFERSA, Mossoró 59625-900, RN, Brazil; (A.M.S.); (A.G.P.); (L.G.P.B.); (S.S.J.M.); (A.F.P.); (M.F.O.)
| | - Ana G. Pereira
- Laboratory of Animal Germplasm Conservation, Department of Animal Sciences, Federal University of Semiarid Region–UFERSA, Mossoró 59625-900, RN, Brazil; (A.M.S.); (A.G.P.); (L.G.P.B.); (S.S.J.M.); (A.F.P.); (M.F.O.)
| | - Luana G. P. Bezerra
- Laboratory of Animal Germplasm Conservation, Department of Animal Sciences, Federal University of Semiarid Region–UFERSA, Mossoró 59625-900, RN, Brazil; (A.M.S.); (A.G.P.); (L.G.P.B.); (S.S.J.M.); (A.F.P.); (M.F.O.)
| | - Samara S. Jerônimo Moreira
- Laboratory of Animal Germplasm Conservation, Department of Animal Sciences, Federal University of Semiarid Region–UFERSA, Mossoró 59625-900, RN, Brazil; (A.M.S.); (A.G.P.); (L.G.P.B.); (S.S.J.M.); (A.F.P.); (M.F.O.)
| | - Alexsandra F. Pereira
- Laboratory of Animal Germplasm Conservation, Department of Animal Sciences, Federal University of Semiarid Region–UFERSA, Mossoró 59625-900, RN, Brazil; (A.M.S.); (A.G.P.); (L.G.P.B.); (S.S.J.M.); (A.F.P.); (M.F.O.)
| | - Moacir F. Oliveira
- Laboratory of Animal Germplasm Conservation, Department of Animal Sciences, Federal University of Semiarid Region–UFERSA, Mossoró 59625-900, RN, Brazil; (A.M.S.); (A.G.P.); (L.G.P.B.); (S.S.J.M.); (A.F.P.); (M.F.O.)
| | - Pierre Comizzoli
- Smithsonian Conservation Biology Institute, National Zoological Park, Veterinary Hospital, Washington, DC 20008, USA;
| | - Alexandre R. Silva
- Laboratory of Animal Germplasm Conservation, Department of Animal Sciences, Federal University of Semiarid Region–UFERSA, Mossoró 59625-900, RN, Brazil; (A.M.S.); (A.G.P.); (L.G.P.B.); (S.S.J.M.); (A.F.P.); (M.F.O.)
- Correspondence:
| |
Collapse
|
17
|
Binsila B, Selvaraju S, Ranjithkumaran R, Archana SS, Krishnappa B, Ghosh SK, Kumar H, Subbarao RB, Arangasamy A, Bhatta R. Current scenario and challenges ahead in application of spermatogonial stem cell technology in livestock. J Assist Reprod Genet 2021; 38:3155-3173. [PMID: 34661801 DOI: 10.1007/s10815-021-02334-7] [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: 01/06/2021] [Accepted: 09/27/2021] [Indexed: 11/28/2022] Open
Abstract
PURPOSE Spermatogonial stem cells (SSCs) are the source for the mature male gamete. SSC technology in humans is mainly focusing on preserving fertility in cancer patients. Whereas in livestock, it is used for mining the factors associated with male fertility. The review discusses the present status of SSC biology, methodologies developed for in vitro culture, and challenges ahead in establishing SSC technology for the propagation of superior germplasm with special reference to livestock. METHOD Published literatures from PubMed and Google Scholar on topics of SSCs isolation, purification, characterization, short and long-term culture of SSCs, stemness maintenance, epigenetic modifications of SSCs, growth factors, and SSC cryopreservation and transplantation were used for the study. RESULT The fine-tuning of SSC isolation and culture conditions with special reference to feeder cells, growth factors, and additives need to be refined for livestock. An insight into the molecular mechanisms involved in maintaining stemness and proliferation of SSCs could facilitate the dissemination of superior germplasm through transplantation and transgenesis. The epigenetic influence on the composition and expression of the biomolecules during in vitro differentiation of cultured cells is essential for sustaining fertility. The development of surrogate males through gene-editing will be historic achievement for the foothold of the SSCs technology. CONCLUSION Detailed studies on the species-specific factors regulating the stemness and differentiation of the SSCs are required for the development of a long-term culture system and in vitro spermatogenesis in livestock. Epigenetic changes in the SSCs during in vitro culture have to be elucidated for the successful application of SSCs for improving the productivity of the animals.
Collapse
Affiliation(s)
- Balakrishnan Binsila
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India.
| | - Sellappan Selvaraju
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Rajan Ranjithkumaran
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Santhanahalli Siddalingappa Archana
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Balaganur Krishnappa
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Subrata Kumar Ghosh
- Animal Reproduction Division, Indian Council of Agricultural Research-Indian Veterinary Research Institute, Izatnagar, 243 122, India
| | - Harendra Kumar
- Animal Reproduction Division, Indian Council of Agricultural Research-Indian Veterinary Research Institute, Izatnagar, 243 122, India
| | - Raghavendra B Subbarao
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Arunachalam Arangasamy
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Raghavendra Bhatta
- Indian council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| |
Collapse
|
18
|
Joshi VB, Behl S, Pittock ST, Arndt CAS, Zhao Y, Khan Z, Granberg CF, Chattha A. Establishment of a Pediatric Ovarian and Testicular Cryopreservation Program for Malignant and Non-Malignant Conditions: The Mayo Clinic Experience. J Pediatr Adolesc Gynecol 2021; 34:673-680. [PMID: 33910089 DOI: 10.1016/j.jpag.2021.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/26/2021] [Accepted: 04/09/2021] [Indexed: 01/15/2023]
Abstract
STUDY OBJECTIVES To describe the structure of a pediatric fertility preservation (FP) program and to share safety and patient satisfaction data. DESIGN The FP program operates under prospective research protocols approved by the Mayo Clinic Institutional Review Board (IRB). SETTING The FP program is a multidisciplinary effort between pediatric gynecology, reproductive endocrinology, pediatric urology, pediatric surgery, and laboratory medicine. PARTICIPANTS The FP program enrolls patients between 0-17 years of age who have been diagnosed with a fertility-threatening condition and/or are scheduled to undergo gonadotoxic treatment. INTERVENTIONS FP is offered in the form of ovarian tissue cryopreservation (OTC) and testicular (TTC) tissue cryopreservation. MAIN OUTCOME MEASURES The outcome measures are the safety of the procedure and results of patient surveys conducted by phone using a standard list of questions to assess attitudes towards FP. RESULTS To date, we have enrolled 38 OTC and 37 TTC patients. The median age (range) of OTC and TTC patients was 11 years (0.83-17 years) and 10 years (0.92-17 years) at the time of enrollment, respectively. Childhood cancers currently represent 88% of the fertility-threatening diagnoses. Meanwhile, patients with non-malignant conditions include those with gender dysphoria, aplastic anemia, and Turner's syndrome. To date, no serious adverse events (SAEs) have been reported following surgery. According to n = 34 one-year follow-ups, 100% of parents felt that FP was a good decision. CONCLUSION Consistent with the literature, our data suggests FP is safe and improves the quality of care provided to pediatric patients for their fertility-threatening diagnoses and/or treatments. TRIAL REGISTRATION NCT02872532, NCT02646384.
Collapse
Affiliation(s)
- Vidhu B Joshi
- Department of Urology, Mayo Clinic, Rochester, Minnesota
| | - Supriya Behl
- Children's Research Center, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Siobhan T Pittock
- Division of Pediatric Endocrinology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Carola A S Arndt
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Yulian Zhao
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Zaraq Khan
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | | | - Asma Chattha
- Division of Pediatric and Adolescent Gynecology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
19
|
Doungkamchan C, Orwig KE. Recent advances: fertility preservation and fertility restoration options for males and females. Fac Rev 2021; 10:55. [PMID: 34195694 PMCID: PMC8204761 DOI: 10.12703/r/10-55] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Fertility preservation is the process of saving gametes, embryos, gonadal tissues and/or gonadal cells for individuals who are at risk of infertility due to disease, medical treatments, age, genetics, or other circumstances. Adult patients have the options to preserve eggs, sperm, or embryos that can be used in the future to produce biologically related offspring with assisted reproductive technologies. These options are not available to all adults or to children who are not yet producing mature eggs or sperm. Gonadal cells/tissues have been frozen for several thousands of those patients worldwide with anticipation that new reproductive technologies will be available in the future. Therefore, the fertility preservation medical and research communities are obligated to responsibly develop next-generation reproductive technologies and translate them into clinical practice. We briefly describe standard options to preserve and restore fertility, but the emphasis of this review is on experimental options, including an assessment of readiness for translation to the human fertility clinic.
Collapse
Affiliation(s)
- Chatchanan Doungkamchan
- Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kyle E Orwig
- Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
20
|
Yan M, Bustos SS, Kuruoglu D, Ciudad P, Forte AJ, Kim EA, Del Corral G, Manrique OJ. Systematic review of fertility preservation options in transgender patients: a guide for plastic surgeons. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:613. [PMID: 33987311 DOI: 10.21037/atm-20-4523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Transgender patients often desire to have biological children. However, their reproductive potential is often negatively impacted by gender affirming surgery (GAS) such as gender confirmation surgery (bottom surgery) and medical hormone therapy. Therefore, counselling patients on fertility preservation options before initiating gender-affirming treatments is prudent to avoid reducing their reproductive potential. A systematic review of English, Spanish, Chinese, French and Turkish languages from 2000 to December 23rd, 2019, using the preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) guidelines, was conducted. The search strategy was designed and conducted by an experienced librarian with input from the study's principle investigator. Fifteen articles that report outcomes of fertility preservation options in transgenders were included. Eight articles described options for transgender women, six reported options for transgender men and one included both transgender women and transgender men. Semen cryopreservation and oocyte cryopreservation are the most common and available methods for fertility preservation in transgenders. Physician awareness of fertility preservation options in transgender patients is crucial to ensure informed discussions regarding reproductive options in the early phase of transition.
Collapse
Affiliation(s)
- Maria Yan
- Division of Plastic and Reconstructive Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Samyd S Bustos
- Division of Plastic and Reconstructive Surgery, Mayo Clinic, Rochester, Minnesota, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Doga Kuruoglu
- Division of Plastic and Reconstructive Surgery, Mayo Clinic, Rochester, Minnesota, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Pedro Ciudad
- Department of Plastic, Reconstructive and Burn Surgery, Arzobispo Loayza National Hospital, Lima, Peru
| | - Antonio J Forte
- Division of Plastic and Reconstructive Surgery, Mayo Clinic Florida Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Jacksonville, USA
| | - Esther A Kim
- Division of Plastic and Reconstructive Surgery, University of California, San Francisco, California, USA
| | - Gabriel Del Corral
- Department of Plastic Surgery, Medstar Georgetown University Hospital, Washington, District of Columbia, USA
| | - Oscar J Manrique
- Division of Plastic and Reconstructive Surgery, Mayo Clinic, Rochester, Minnesota, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
21
|
Strategies for cryopreservation of testicular cells and tissues in cancer and genetic diseases. Cell Tissue Res 2021; 385:1-19. [PMID: 33791878 DOI: 10.1007/s00441-021-03437-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/18/2021] [Indexed: 12/15/2022]
Abstract
Cryopreservation of testicular cells and tissues is useful for the preservation and restoration of fertility in pre-pubertal males expecting gonadotoxic treatment for cancer and genetic diseases causing impaired spermatogenesis. A number of freezing and vitrification protocols have thus been tried and variable results have been reported in terms of cell viability spermatogenesis progression and the production of fertile spermatozoa. A few studies have also reported the production of live offspring from cryopreserved testicular stem cells and tissues in rodents but their replication in large animals and human have been lacking. Advancement in in vitro spermatogenesis system has improved the possibility of producing fertile spermatozoa from the cryopreserved testis and has reduced the dependency on transplantation. This review provides an update on various cryopreservation strategies for fertility preservation in males expecting gonadotoxic treatment. It also discusses various methods of assessing and ameliorating cryoinjuries. Newer developments on in vitro spermatogenesis and testicular tissue engineering for in vitro sperm production from cryopreserved SSCs and testicular tissue are also discussed.
Collapse
|
22
|
Maria da Silva A, Pereira AG, Brasil AV, Macedo LB, Souza-Junior J, Bezerra de Moura CE, Pereira AF, Franco de Oliveira M, Comizzoli P, Silva AR. Influence of freezing techniques and glycerol-based cryoprotectant combinations on the survival of testicular tissues from adult collared peccaries. Theriogenology 2021; 167:111-119. [PMID: 33813051 DOI: 10.1016/j.theriogenology.2021.03.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 01/23/2023]
Abstract
The objective of the study was to evaluate the effects of different cryopreservation techniques including glycerol-based cryoprotectant combinations on the structure and viability of testicular tissues from adult collared peccaries. Tissue biopsies (3.0 mm³) from 5 different individuals were allocated to 10 different groups: fresh control; slow freezing (SF), conventional vitrification (CV), or solid-surface vitrification (SSV); each of them using three different combinations of cryoprotectants [dimethyl sulfoxide (DMSO) + ethylene glycol (EG); DMSO + Glycerol; and EG + Glycerol]. After thawing/warming, samples were evaluated for histomorphology, viability, proliferative capacity potential, and DNA integrity. Most effective preservation of testicular histomorphology was achieved using SF and CV with DMSO + EG. However, the use of glycerol-based cryoprotectant combinations increased the occurrence of tubular cell swelling, tubular cell loss and shrinkage from the basal membrane. Cell viability was comparable among cryopreservation methods and cryoprotectant combinations. Regarding cell proliferative capacity, the use of SF with EG + Glycerol and SSV with DMSO + Glycerol impaired the conservation of spermatogonia proliferative potential compared to other treatments. Moreover, CV with DMSO + EG was better than SF with EG + Glycerol for Sertoli cell proliferation potential. Regarding DNA integrity, less damage occurred when using SF with DMSO + EG while more fragmentations were observed when using CV with EG + Glycerol or DMSO + Glycerol as well as SSV with EG + Glycerol or DMSO + Glycerol. In sum, SF and CV appeared to be the most suitable methods for the cryopreservation of adult peccary testicular tissues. Additionally, the use of glycerol-based cryoprotectant combinations did not improve testicular tissues preservation with DMSO + EG being the most efficient option.
Collapse
Affiliation(s)
- Andréia Maria da Silva
- Laboratory of Animal Germplasm Conservation, Federal Rural University of Semi-Arid - UFERSA, Mossoró, RN, Brazil
| | - Ana Gloria Pereira
- Laboratory of Animal Germplasm Conservation, Federal Rural University of Semi-Arid - UFERSA, Mossoró, RN, Brazil
| | - Andreza Vieira Brasil
- Laboratory of Animal Germplasm Conservation, Federal Rural University of Semi-Arid - UFERSA, Mossoró, RN, Brazil
| | | | - João Souza-Junior
- Laboratory of Animal Germplasm Conservation, Federal Rural University of Semi-Arid - UFERSA, Mossoró, RN, Brazil
| | | | | | | | - Pierre Comizzoli
- Smithsonian Conservation Biology Institute, National Zoological Park, Veterinary Hospital, Washington, DC, USA
| | - Alexandre Rodrigues Silva
- Laboratory of Animal Germplasm Conservation, Federal Rural University of Semi-Arid - UFERSA, Mossoró, RN, Brazil.
| |
Collapse
|
23
|
Andrae CS, Oliveira ECS, Ferraz MAMM, Nagashima JB. Cryopreservation of grey wolf (Canis lupus) testicular tissue. Cryobiology 2021; 100:173-179. [PMID: 33482146 DOI: 10.1016/j.cryobiol.2021.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/30/2020] [Accepted: 01/12/2021] [Indexed: 12/26/2022]
Abstract
Development of genomic preservation technologies for canids, especially for seasonally breeding species like the grey wolf (Canis lupus), is needed in advance of growing species conservation concerns. Here, we evaluated the efficacy of two cryopreservation protocols - needle immersion vitrification (NIV) and slow freezing (SF) on grey wolf (n = 7) testicular tissue morphology. NIV samples were equilibrated in a 7.5% v/v dimethyl sulfoxide (DMSO or Me2SO) + 7.5% ethylene glycol (EG) solution in minimum essential medium with 20% FBS for 10 min at 4 °C, then exposed to 15% DMSO + 15% EG + 0.5 M sucrose for 10 min at 4 °C before plunging into liquid nitrogen. For slow freezing, we assessed two cryoprotectant (CPA) strategies, DMSO, 15% v/v alone (SF-D) or 7.5% EG + 7.5% DMSO (SF-ED). Following thawing, there were no significant differences in seminiferous tubule area among treatment groups, although all cryopreserved tissues displayed reduced tubule size compared with fresh controls and increased apoptosis, the latter reaching significance for SF-D treated tissues. Slow freezing improved maintenance of testis architecture, with minimal detachment of seminiferous tubule basement membranes post-thaw. Spermatogonia densities were reduced in NIV tissues compared with fresh, with no differences in spermatocyte, spermatid, or Sertoli cell counts, or germ cell marker DDX4+ cell densities among groups. In sum, we conclude that slow freezing better maintained morphology of cryopreserved testicular tissues compared with needle vitrification with 15% each DMSO and EG and 0.5 M sucrose, and that DMSO + EG combination SF supports cell viability. This represents a first step in the development of male gonadal tissue preservation strategies for the grey wolf.
Collapse
Affiliation(s)
- Christopher S Andrae
- Smithsonian-Mason School of Conservation, George Mason University, 1500 Remount Rd., Front Royal, VA, 22630, USA
| | - Erika C S Oliveira
- University of Virginia, Department of Cell Biology, 200 Jeanette Lancaster Way, Charlottesville, VA, 22903, USA
| | - Marcia A M M Ferraz
- Smithsonian Conservation Biology Institute, 1500 Remount Rd., Front Royal, VA, 22630, USA; Gene Center, Ludwig-Maximilians University, Feodor-Lynen Str. 25, D-81377, Munich, Germany
| | - Jennifer B Nagashima
- Smithsonian Conservation Biology Institute, 1500 Remount Rd., Front Royal, VA, 22630, USA.
| |
Collapse
|
24
|
Kaneko H, Kikuchi K, Men NT, Dang-Nguyen TQ, Oyadomari M, Touma S, Suzuki N, Katagiri Y. Embryo production by intracytoplasmic injection of sperm retrieved from neonatal testicular tissue of Agu pigs after cryopreservation and grafting into nude mice. Anim Sci J 2021; 91:e13479. [PMID: 33331680 DOI: 10.1111/asj.13479] [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: 08/14/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 11/27/2022]
Abstract
The Agu is the only indigenous pig breed in Japan but its population is very small. In order to estimate the efficacy of testicular xenografting for the conservation of Agu pigs, we investigated whether neonatal testicular fragments would acquire the capacity to produce sperm after they had been cryopreserved and grafted into nude mice. Although on day 180 (day 0 = xenografting), grafts showed a low proportion of seminiferous tubule cross-sections containing sperm (0.1 ± 0.1%, mean ± SEM for four mice), the proportion reached 36.9 ± 16.7% (n = 4 mice) by day 240. When single sperm obtained on day 240 was injected into individual porcine oocytes, 28.2% of the oocytes were found to contain one male and one female pronuclei with the second polar body. Moreover, the blastocyst formation rate after injection of the xenogeneic sperm was 28.4%, whereas that in the absence of sperm injection (attributable to parthenogenesis) was 13.3%. These findings suggest that more than half of the blastocysts resulted from fertilization. Thus, testicular xenografting could assist the conservation of Agu pigs by salvaging germ cells present in neonatal testes even after cryopreservation.
Collapse
Affiliation(s)
- Hiroyuki Kaneko
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Japan
| | - Kazuhiro Kikuchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Japan.,The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Nguyen Thi Men
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Japan
| | - Thanh Quang Dang-Nguyen
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Japan
| | - Motoharu Oyadomari
- Okinawa Prefectural Livestock and Grassland Research Center, Nakijin, Japan
| | - Shihei Touma
- Okinawa Prefectural Livestock and Grassland Research Center, Nakijin, Japan
| | - Naoto Suzuki
- Okinawa Prefectural Livestock and Grassland Research Center, Nakijin, Japan
| | - Yoshito Katagiri
- Okinawa Prefectural Livestock and Grassland Research Center, Nakijin, Japan
| |
Collapse
|
25
|
Wyns C, Kanbar M, Giudice MG, Poels J. Fertility preservation for prepubertal boys: lessons learned from the past and update on remaining challenges towards clinical translation. Hum Reprod Update 2020; 27:433-459. [PMID: 33326572 DOI: 10.1093/humupd/dmaa050] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/25/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Childhood cancer incidence and survivorship are both on the rise. However, many lifesaving treatments threaten the prepubertal testis. Cryopreservation of immature testicular tissue (ITT), containing spermatogonial stem cells (SSCs), as a fertility preservation (FP) option for this population is increasingly proposed worldwide. Recent achievements notably the birth of non-human primate (NHP) progeny using sperm developed in frozen-thawed ITT autografts has given proof of principle of the reproductive potential of banked ITT. Outlining the current state of the art on FP for prepubertal boys is crucial as some of the boys who have cryopreserved ITT since the early 2000s are now in their reproductive age and are already seeking answers with regards to their fertility. OBJECTIVE AND RATIONALE In the light of past decade achievements and observations, this review aims to provide insight into relevant questions for clinicians involved in FP programmes. Have the indications for FP for prepubertal boys changed over time? What is key for patient counselling and ITT sampling based on the latest achievements in animals and research performed with human ITT? How far are we from clinical application of methods to restore reproductive capacity with cryostored ITT? SEARCH METHODS An extensive search for articles published in English or French since January 2010 to June 2020 using keywords relevant to the topic of FP for prepubertal boys was made in the MEDLINE database through PubMed. Original articles on fertility preservation with emphasis on those involving prepubertal testicular tissue, as well as comprehensive and systematic reviews were included. Papers with redundancy of information or with an absence of a relevant link for future clinical application were excluded. Papers on alternative sources of stem cells besides SSCs were excluded. OUTCOMES Preliminary follow-up data indicate that around 27% of boys who have undergone testicular sampling as an FP measure have proved azoospermic and must therefore solely rely on their cryostored ITT to ensure biologic parenthood. Auto-transplantation of ITT appears to be the first technique that could enter pilot clinical trials but should be restricted to tissue free of malignant cells. While in vitro spermatogenesis circumvents the risk linked to cancer cell contamination and has led to offspring in mice, complete spermatogenesis has not been achieved with human ITT. However, generation of haploid germ cells paves the way to further studies aimed at completing the final maturation of germ cells and increasing the efficiency of the processes. WIDER IMPLICATIONS Despite all the research done to date, FP for prepubertal boys remains a relatively young field and is often challenging to healthcare providers, patients and parents. As cryopreservation of ITT is now likely to expand further, it is important not only to acknowledge some of the research questions raised on the topic, e.g. the epigenetic and genetic integrity of gametes derived from strategies to restore fertility with banked ITT but also to provide healthcare professionals worldwide with updated knowledge to launch proper multicollaborative care pathways in the field and address clinical issues that will come-up when aiming for the child's best interest.
Collapse
Affiliation(s)
- Christine Wyns
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc Kanbar
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Maria Grazia Giudice
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jonathan Poels
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
26
|
Sabetkish S, Kajbafzadeh AM, Sabetkish N. Recellularization of testicular feminization testis in C57bl6 as a natural bioreactor for creation of cellularized seminiferous tubules: an experimental study. Cell Tissue Bank 2020; 22:287-295. [PMID: 33146808 DOI: 10.1007/s10561-020-09877-6] [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: 07/06/2019] [Accepted: 10/22/2020] [Indexed: 11/24/2022]
Abstract
We determined histological aspects of implanted human decellularized testicular matrix (DTM) in C57BL6 as a primitive step for further testis tissue engineering. A total of 4 immature human testicles were obtained after bilateral orchiectomy from patients with testicular feminization syndrome. The optimal decellularization protocol was determined and the efficacy of decellularization was evaluated in two of the testicles. The remaining scaffolds were cut in 3 × 3 mm3 pieces and implanted between the tight muscles in 32 C57BL6. Biopsies were taken at 2, 4, 8, and 24 weeks postoperatively and stained with PLZF, protamine, and tekt1 markers. Histological examination of DTMs confirmed complete absence of nuclear remnants and preservation of the extracellular matrix. Successful cell seeding was observed in all follow-ups confirmed by H&E and IHC staining that increased continuously during the whole study. Interestingly, spermatogonial stem-like cells were observed on decellularized implants that were well differentiated during the follow-ups. Natural bioreactors may provide a good cell source for testes tissue regeneration. This technique may provide testis bioscaffold as a three-dimensional platform and further successful cell seeding to produce a functional testis. This novel technique may be beneficial for patients who require testicular supplementation.
Collapse
Affiliation(s)
- Shabnam Sabetkish
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419433151, Iran
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419433151, Iran.
| | - Nastaran Sabetkish
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419433151, Iran
| |
Collapse
|
27
|
|
28
|
Silva AMD, Pereira AF, Comizzoli P, Silva AR. Cryopreservation and Culture of Testicular Tissues: An Essential Tool for Biodiversity Preservation. Biopreserv Biobank 2020; 18:235-243. [PMID: 32282240 DOI: 10.1089/bio.2020.0010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Systematic cryo-banking of reproductive tissues could enhance reproductive management and ensure sustainability of rare mammalian genotypes. Testicular tissues contain a vast number of germ cells, including at early stages (spermatogonia and spermatocytes), that can potentially develop into viable spermatozoa after grafting or culture in vitro, and the resulting sperm cells then can be used for assisted reproductive techniques. The objective of this review was to describe current advances, limitations, and perspectives related to the use of testicular tissue preservation as a strategy for the conservation of male fertility. Testes can be obtained from mature or prepubertal individuals, immediately postmortem or by orchiectomy, but testicular biopsies could also be an alternative to collect samples from living individuals. Testicular fragments can be then cryopreserved by using slow or ultra-rapid freezing, or even vitrification methods. The composition of cryopreservation media can vary according to species-specific characteristics, especially regarding the cryoprotectant type and concentration. Finally, spermatozoa have been usually obtained after xenografting of testicular fragments into severely immunodeficient mice, while this method still has to be optimized after in vitro culture conditions.
Collapse
Affiliation(s)
- Andréia Maria da Silva
- Laboratory of Animal Germplasm Conservation, Federal Rural University of Semi-Arid, Mossoró, Brazil
| | | | - Pierre Comizzoli
- Smithsonian Conservation Biology Institute, National Zoological Park, Veterinary Hospital, Washington, District of Columbia, USA
| | | |
Collapse
|
29
|
Gargus ES, Rogers HB, McKinnon KE, Edmonds ME, Woodruff TK. Engineered reproductive tissues. Nat Biomed Eng 2020; 4:381-393. [PMID: 32251392 PMCID: PMC7416444 DOI: 10.1038/s41551-020-0525-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 02/05/2020] [Indexed: 12/14/2022]
Abstract
Engineered male and female biomimetic reproductive tissues are being developed as autonomous in vitro units or as integrated multi-organ in vitro systems to support germ cell and embryo function, and to display characteristic endocrine phenotypic patterns, such as the 28-day human ovulatory cycle. In this Review, we summarize how engineered reproductive tissues facilitate research in reproductive biology, and overview strategies for making engineered reproductive tissues that might eventually allow the restoration of reproductive capacity in patients.
Collapse
Affiliation(s)
- Emma S Gargus
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hunter B Rogers
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kelly E McKinnon
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maxwell E Edmonds
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Teresa K Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
30
|
Gül M, Dong L, Wang D, Diri MA, Andersen CY. Surrogate testes: Allogeneic spermatogonial stem cell transplantation within an encapsulation device may restore male fertility. Med Hypotheses 2020; 139:109634. [PMID: 32087491 DOI: 10.1016/j.mehy.2020.109634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/08/2020] [Accepted: 02/13/2020] [Indexed: 01/15/2023]
Abstract
Toxic insult to the gonads by chemotherapy or radiotherapy can lead to permanent infertility. It's an important health concern because each year more than 4000 male patients are at risk of azoospermia in the United States due to gonadotoxicity of the regimens used. There are also several benign/genetic diseases whose natural course can result in infertility without gonadotoxic therapy. Considering the fact that most of these people are cured and survive with the advent of modern medicine, infertility is related to serious psychological and relationship implications and parenthood is a significant issue for those patients. Semen cryopreservation option is available for postpubertal adolescent and adult men, while children do not have this storing option since they do not have mature spermatozoa. However, their testes contain spermatogonial stem cells (SSCs), which are initiators of spermatogenesis. Promising findings in animal studies and human cell lines have encouraged scientists that SSCs may be hope for restoring fertility option of patients who cannot produce functional sperm and who have no other choice to preserve their future fertility. For this reason, several centers around the world already began to collect and cryopreserve testicular tissue or cells with anticipation that SSC-based therapies will be available in the near future; however, an optimal transplantation design in humans is yet to be developed. Here we propose an allogeneic testicular stem cell transplantation with an encapsulation device to restore fertility in patients with infertility. We endeavor to discuss the reliability of this method with the current literature and bring the evidence on its feasibility.
Collapse
Affiliation(s)
- Murat Gül
- Laboratory of Reproductive Biology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark; Department of Urology, Selcuk University School of Medicine, 42250 Konya, Turkey.
| | - Lihua Dong
- Laboratory of Reproductive Biology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Danyang Wang
- Laboratory of Reproductive Biology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Mehmet Akif Diri
- Department of Urology, Aksaray University School of Medicine, 68100 Aksaray, Turkey
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| |
Collapse
|
31
|
Abstract
This chapter highlights the importance of reproductive technologies that are applied to porcine breeds. Nowadays the porcine industry, part of a high technological and specialized sector, offers high-quality protein food. The development of the swine industry is founded in the development of breeding/genetics, nutrition, animal husbandry, and animal health. The implementation of reproductive technologies in swine has conducted to levels of productivity never reached before. In addition, the pig is becoming an important species for biomedicine. The generation of pig models for human disease, xenotransplantation, or production of therapeutic proteins for human medicine has in fact generated a growing field of interest.
Collapse
|
32
|
Fertility preservation in patients undergoing gonadotoxic therapy or gonadectomy: a committee opinion. Fertil Steril 2019; 112:1022-1033. [DOI: 10.1016/j.fertnstert.2019.09.013] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 02/08/2023]
|
33
|
da Silva AM, Bezerra LGP, Praxedes ECG, Moreira SSJ, de Souza CMP, de Oliveira MF, Pereira AF, Comizzoli P, Silva AR. Combination of intracellular cryoprotectants preserves the structure and the cells proliferative capacity potential of adult collared peccary testicular tissue subjected to solid surface vitrification. Cryobiology 2019; 91:53-60. [DOI: 10.1016/j.cryobiol.2019.10.199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/22/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022]
|
34
|
Gauthier-Fisher A, Kauffman A, Librach CL. Potential use of stem cells for fertility preservation. Andrology 2019; 8:862-878. [PMID: 31560823 DOI: 10.1111/andr.12713] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Infertility and gonadal dysfunction can result from gonadotoxic therapies, environmental exposures, aging, or genetic conditions. In men, non-obstructive azoospermia (NOA) results from defects in the spermatogenic process that can be attributed to spermatogonial stem cells (SSC) or their niche, or both. While assisted reproductive technologies and sperm banking can enable fertility preservation (FP) in men of reproductive age who are at risk for infertility, FP for pre-pubertal patients remains experimental. Therapeutic options for NOA are limited. The rapid advance of stem cell research and of gene editing technologies could enable new FP options for these patients. Induced pluripotent stem cells (iPSC), SSC, and testicular niche cells, as well as mesenchymal stromal cells (aka medicinal signaling cells, MSCs), have been investigated for their potential use in male FP strategies. OBJECTIVE Here, we review the benefits and challenges for three types of stem cell-based approaches under investigation for male FP, focusing on the role that promising sources of MSC derived from human umbilical cord, specifically human umbilical cord perivascular cells (HUCPVC), could fulfill. These approaches are as follows: 1. isolation and ex vivo expansion of autologous SSC for in vivo transplantation or in vitro spermatogenesis; 2. in vitro differentiation toward germ cell and testicular somatic cell lineages using autologous SSC, or stem cells such iPSC or MSC; and 3. protection or regeneration of the spermatogenic niche after gonadotoxic insults in vivo. CONCLUSION Our studies suggest that HUCPVC are promising sources of cells that could be utilized in multiple aspects of male FP strategies.
Collapse
Affiliation(s)
| | - A Kauffman
- CReATe Fertility Centre, Toronto, ON, Canada
| | - C L Librach
- CReATe Fertility Centre, Toronto, ON, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Gynecology, Women's College Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
35
|
Oliver E, Stukenborg JB. Rebuilding the human testis in vitro. Andrology 2019; 8:825-834. [PMID: 31539453 PMCID: PMC7496374 DOI: 10.1111/andr.12710] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/21/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022]
Abstract
Increasing rates of male infertility have led to a greater need for relevant model systems to gain further insight into male fertility and its failings. Spermatogenesis and hormone production occur within distinct regions of the testis. Defined by specialized architecture and a diverse population of cell types, it is no surprise that disruption of this highly organized microenvironment can lead to infertility. To date, no robust in vitro system has facilitated full spermatogenesis resulting in the production of fertilization‐competent human spermatozoa. Here, we review a selection of current in vitro systems available for modelling the human testis microenvironment with focus on the progression of spermatogenesis and recapitulation of the testis microenvironment.
Collapse
Affiliation(s)
- E Oliver
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - J-B Stukenborg
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
36
|
Gene expression analysis of ovine prepubertal testicular tissue vitrified with a novel cryodevice (E.Vit). J Assist Reprod Genet 2019; 36:2145-2154. [PMID: 31414315 DOI: 10.1007/s10815-019-01559-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/06/2019] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Testicular tissue cryopreservation prior to gonadotoxic therapies is a method to preserve fertility in children. However, the technique still requires development, especially when the tissue is immature and rather susceptible to stress derived from in vitro manipulation. This study aimed to investigate the effects of vitrification with a new cryodevice (E.Vit) on cell membrane integrity and gene expression of prepubertal testicular tissue in the ovine model. METHODS Pieces of immature testicular tissue (1 mm3) were inserted into "E.Vit" devices and vitrified with a two-step protocol. After warming, tissues were cultured in vitro and cell membrane integrity was assessed after 0, 2, and 24 h by trypan blue exclusion test. Controls consisted of non-vitrified tissue analyzed after 0, 2, and 24 h in vitro culture (IVC). Expression of genes involved in transcriptional stress response (BAX, SOD1, CIRBP, HSP90AB1), cell proliferation (KIF11), and germ- (ZBDB16, TERT, POU5F1, KIT) and somatic- (AR, FSHR, STAR) cell specific markers was evaluated 2 and 24 h after warming. RESULTS Post-warming trypan blue staining showed the survival of most cells, although membrane integrity immediately after warming (66.00% ± 4.73) or after 2 h IVC (59.67% ± 4.18) was significantly lower than controls (C0h 89.67% ± 1.45). Extended post-warming IVC (24 h) caused an additional decrease to 31% ± 3.46 (P < 0.05). Germ- and somatic-cell specific markers showed the survival of both cell types after cryopreservation and IVC. All genes were affected by cryopreservation and/or IVC, and moderate stress conditions were indicated by transcriptional stress response. CONCLUSIONS Vitrification with the cryodevice E.Vit is a promising strategy to cryopreserve prepubertal testicular tissue.
Collapse
|
37
|
Vermeulen M, Giudice MG, Del Vento F, Wyns C. Role of stem cells in fertility preservation: current insights. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2019; 12:27-48. [PMID: 31496751 PMCID: PMC6689135 DOI: 10.2147/sccaa.s178490] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/24/2019] [Indexed: 12/11/2022]
Abstract
While improvements made in the field of cancer therapy allow high survival rates, gonadotoxicity of chemo- and radiotherapy can lead to infertility in male and female pre- and postpubertal patients. Clinical options to preserve fertility before starting gonadotoxic therapies by cryopreserving sperm or oocytes for future use with assisted reproductive technology (ART) are now applied worldwide. Cryopreservation of pre- and postpubertal ovarian tissue containing primordial follicles, though still considered experimental, has already led to the birth of healthy babies after autotransplantation and is performed in an increasing number of centers. For prepubertal boys who do not produce gametes ready for fertilization, cryopreservation of immature testicular tissue (ITT) containing spermatogonial stem cells may be proposed as an experimental strategy with the aim of restoring fertility. Based on achievements in nonhuman primates, autotransplantation of ITT or testicular cell suspensions appears promising to restore fertility of young cancer survivors. So far, whether in two- or three-dimensional culture systems, in vitro maturation of immature male and female gonadal cells or tissue has not demonstrated a capacity to produce safe gametes for ART. Recently, primordial germ cells have been generated from embryonic and induced pluripotent stem cells, but further investigations regarding efficiency and safety are needed. Transplantation of mesenchymal stem cells to improve the vascularization of gonadal tissue grafts, increase the colonization of transplanted cells, and restore the damaged somatic compartment could overcome the current limitations encountered with transplantation.
Collapse
Affiliation(s)
- Maxime Vermeulen
- Gynecology-Andrology Research Unit, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, 1200, Belgium
| | - Maria-Grazia Giudice
- Gynecology-Andrology Research Unit, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, 1200, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels 1200, Belgium
| | - Federico Del Vento
- Gynecology-Andrology Research Unit, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, 1200, Belgium
| | - Christine Wyns
- Gynecology-Andrology Research Unit, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, 1200, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels 1200, Belgium
| |
Collapse
|
38
|
Porter LH, Lawrence MG, Wang H, Clark AK, Bakshi A, Obinata D, Goode D, Papargiris M, Clouston D, Ryan A, Norden S, Corey E, Nelson PS, Isaacs JT, Grummet J, Kourambas J, Sandhu S, Murphy DG, Pook D, Frydenberg M, Taylor RA, Risbridger GP. Establishing a cryopreservation protocol for patient-derived xenografts of prostate cancer. Prostate 2019; 79:1326-1337. [PMID: 31212368 DOI: 10.1002/pros.23839] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/08/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Serially transplantable patient-derived xenografts (PDXs) are invaluable preclinical models for studying tumor biology and evaluating therapeutic agents. As these models are challenging to establish from prostate cancer specimens, the ability to preserve them through cryopreservation has several advantages for ongoing research. Despite this, there is still uncertainty about the ability to cryopreserve PDXs of prostate cancer. This study compared three different cryopreservation protocols to identify a method that can be used to reproducibly cryopreserve a diverse cohort of prostate cancer PDX models. METHODS One serially transplantable prostate cancer PDX from the Melbourne Urological Research Alliance cohort was used to compare three cryopreservation protocols: slow freezing in fetal calf serum (FCS) with 10% dimethyl sulfoxide (DMSO), FCS with 10% DMSO supplemented with the Rho-associated kinase (ROCK) inhibitor Y-27632 and vitrification. The efficiency of the slow freezing protocols was then assessed in 17 additional prostate cancer PDXs. Following cryopreservation, PDXs were re-established in host mice that were either intact and supplemented with testosterone or castrated. Graft take rate, tumor growth, histological features, and transcriptome profiles before and after cryopreservation were compared. RESULTS Slow freezing maintained the viability and histological features of prostate cancer PDXs, and the addition of a ROCK inhibitor increased their growth following cryopreservation. Using the slow freezing method, we re-established 100% of PDXs grown in either testosterone-supplemented or castrated host mice. Importantly, the long-term tumor growth rate and transcriptome profile were maintained following cryopreservation. CONCLUSION This study has identified a protocol to reliably cryopreserve and re-establish a diverse cohort of serially transplantable PDXs of prostate cancer. This study has the potential to significantly improve the practicality of maintaining PDX models. Cryopreservation may also increase the accessibility of these important resources and provide new opportunities for preclinical studies on a broader spectrum of prostate tumors.
Collapse
Affiliation(s)
- Laura H Porter
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
| | - Mitchell G Lawrence
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
- Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - Hong Wang
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
| | - Ashlee K Clark
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
| | - Andrew Bakshi
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
- Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Daisuke Obinata
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
| | - David Goode
- Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Melissa Papargiris
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
- Australian Prostate Cancer Bioresource, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Victoria Node, Clayton, Victoria, Australia
| | | | - Andrew Ryan
- TissuPath, Mount Waverley, Victoria, Australia
| | - Sam Norden
- TissuPath, Mount Waverley, Victoria, Australia
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington
| | - Peter S Nelson
- Department of Urology, University of Washington, Seattle, Washington
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Pathology, University of Washington, Seattle, Washington
| | - John T Isaacs
- Department of Oncology, Prostate Cancer Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jeremy Grummet
- Department of Surgery, Central Clinical School, Monash University, Clayton, Victoria, Australia
- Epworth Healthcare, Richmond, Victoria, Australia
| | - John Kourambas
- Department of Medicine, Monash Health, Casey Hospital, Berwick, Victoria, Australia
| | - Shahneen Sandhu
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Victoria, Australia
- Cancer Tissue Collection After Death (CASCADE) Program, Peter MacCallum Cancer Centre, University of Melbourne, Victoria, Australia
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
- Epworth Healthcare, Richmond, Victoria, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, University of Melbourne, Victoria, Australia
| | - David Pook
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
- Medical Oncology, Monash Health, Clayton, Victoria, Australia
| | - Mark Frydenberg
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
- Department of Surgery, Central Clinical School, Monash University, Clayton, Victoria, Australia
- Epworth Healthcare, Richmond, Victoria, Australia
- Australian Urology Associates, Melbourne, Victoria, Australia
| | - Renea A Taylor
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
- Department of Physiology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
| | - Gail P Risbridger
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, Victoria, Australia
- Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
39
|
Fayomi AP, Peters K, Sukhwani M, Valli-Pulaski H, Shetty G, Meistrich ML, Houser L, Robertson N, Roberts V, Ramsey C, Hanna C, Hennebold JD, Dobrinski I, Orwig KE. Autologous grafting of cryopreserved prepubertal rhesus testis produces sperm and offspring. Science 2019; 363:1314-1319. [PMID: 30898927 PMCID: PMC6598202 DOI: 10.1126/science.aav2914] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 02/13/2019] [Indexed: 12/11/2022]
Abstract
Testicular tissue cryopreservation is an experimental method to preserve the fertility of prepubertal patients before they initiate gonadotoxic therapies for cancer or other conditions. Here we provide the proof of principle that cryopreserved prepubertal testicular tissues can be autologously grafted under the back skin or scrotal skin of castrated pubertal rhesus macaques and matured to produce functional sperm. During the 8- to 12-month observation period, grafts grew and produced testosterone. Complete spermatogenesis was confirmed in all grafts at the time of recovery. Graft-derived sperm were competent to fertilize rhesus oocytes, leading to preimplantation embryo development, pregnancy, and the birth of a healthy female baby. Pending the demonstration that similar results are obtained in noncastrated recipients, testicular tissue grafting may be applied in the clinic.
Collapse
Affiliation(s)
- Adetunji P Fayomi
- Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Karen Peters
- Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Meena Sukhwani
- Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hanna Valli-Pulaski
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Gunapala Shetty
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marvin L Meistrich
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lisa Houser
- Assisted Reproductive Technology Core, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Nicola Robertson
- Assisted Reproductive Technology Core, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Victoria Roberts
- Assisted Reproductive Technology Core, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Cathy Ramsey
- Assisted Reproductive Technology Core, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Carol Hanna
- Assisted Reproductive Technology Core, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Jon D Hennebold
- Assisted Reproductive Technology Core, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Ina Dobrinski
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kyle E Orwig
- Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
40
|
Altered hormonal milieu and dysregulated protein expression can cause spermatogenic arrest in ectopic xenografted immature rat testis. Sci Rep 2019; 9:4036. [PMID: 30858478 PMCID: PMC6411886 DOI: 10.1038/s41598-019-40662-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/20/2019] [Indexed: 01/15/2023] Open
Abstract
Testis tissue xenografting complemented with cryopreservation is a feasible technique for fertility preservation in children with malignancy receiving gonadotoxic therapy and for endangered species with high neonatal mortality rate. However, xenografted testis of human and most endangered species are known to undergo spermatogenic arrest. In this study, we xenografted immature rat testis onto immunodeficient male mice to investigate the plausible underlying causes of spermatogenic arrest. Histological analysis of xenografted testes collected 8-wk post-grafting showed incomplete spermatogenesis with pachytene-stage spermatocytes as the most advanced germ cells. Although the levels of serum luteinizing hormone and testosterone were normal in recipient mice, those of follicle stimulating hormone (FSH) were significantly high, and specific receptors of FSH were absent in the xenografts. The xenografts demonstrated dysregulated expression of Sertoli cell-transcriptional regulators (WT1 and SOX9) and secretory proteins (SCF and GDNF). In conclusion, results from our study suggested that an altered hormonal milieu in recipients and dysregulated protein expression in xenografts could be a potential cause of spermatogenic arrest in xenografted immature rat testis. Further stereological analysis of xenografts can demonstrate precise cellular composition of xenografts to decipher interactions between germ and somatic cells to better understand spermatogenic arrest in xenografted testis.
Collapse
|
41
|
Kaneko H, Kikuchi K, Men NT, Noguchi J. Embryo production by intracytoplasmic injection of sperm retrieved from Meishan neonatal testicular tissue cryopreserved and grafted into nude mice. Anim Sci J 2018; 90:158-166. [PMID: 30523649 PMCID: PMC6587828 DOI: 10.1111/asj.13138] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/13/2018] [Accepted: 10/17/2018] [Indexed: 11/29/2022]
Abstract
Testicular xenografting, combined with cryopreservation can assist conservation of the genetic diversity of indigenous pigs by salvaging germ cells from their neonatal testes. Using Meishan male piglets as an example, we examined whether testicular tissue would acquire the ability to produce sperm after cryopreservation and grafting into nude mice (MS group). For comparison, testicular tissue from neonatal Western crossbreed male piglets was used (WC group). Sixty days after xenografting (day 0 = grafting), MS grafts had already developed seminiferous tubules containing sperm, whereas in the WC grafts, sperm first appeared on day 120. The proportion of tubules containing spermatids and sperm was higher in the MS group than in the WC group between days 90 and 120. Moreover, in vitro‐matured porcine oocytes injected with a single sperm obtained from the MS group on day 180 developed to the blastocyst stage. The blastocyst formation rate after injection of the xenogeneic sperm was 14.6%, whereas the ratio in the absence of such injection (attributable to parthenogenesis) was 6.7%. Thus, cryopreserved Meishan testicular tissue acquired spermatogenic activity in host mice 60 days earlier than Western crossbreed tissue. Such xenogeneic sperm are likely capable of generating blastocysts in vitro.
Collapse
Affiliation(s)
- Hiroyuki Kaneko
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Kazuhiro Kikuchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan.,The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Nguyen Thi Men
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Junko Noguchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| |
Collapse
|
42
|
Benvenutti L, Salvador RA, Til D, Senn AP, Tames DR, Amaral NLL, Amaral VLL. Wistar rats immature testicular tissue vitrification and heterotopic grafting. JBRA Assist Reprod 2018; 22:167-173. [PMID: 29693963 PMCID: PMC6106629 DOI: 10.5935/1518-0557.20180023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objective To evaluate the efficiency of two vitrification protocols for rat immature
testicular tissue and heterotopic transplantation. Methods Twenty-four pre-pubertal Wistar rats were divided into three groups (n=8).
After orchiectomy, testicular fragments (3mm) from Groups 1 and 2 were
vitrified with different cryoprotectant concentration solutions, using
sterile inoculation loops as support. After warming up, the fragments were
submitted to cell viability assessment by Trypan blue and histological
evaluation. Vitrified (Groups 1 and 2) and fresh (Group 3) fragments were
grafted to the animals periauricular region. After 8 weeks of grafting, the
implant site was histologically analyzed. Results The viability recovery rate from Group 1 (72.09%) was higher
(p=0.02) than that from Group 2 (59.19%). Histological
analysis showed similar tubular integrity between fresh fragments from
Groups 1 and 3. Group 2 samples presented lower tubular integrity. We ran
histological analyses in the grafts from the Groups. In all groups, it was
possible to see the implant site, however, no fragment of testicular tissue
or signs of inflammation were histologically found in most samples from
Groups 1 and 3. In one sample from Group 2, we found degenerated
seminiferous tubules with necrosis and signs of an inflammatory process. In
another sample from Group 2, we found seminiferous tubules in the implant
site. Conclusion The vitrification of pre-pubertal testicular tissue of rats showed little
damage to cell viability through histological analysis when we used
cryoprotectants in a lower concentration. Heterotopic transplantation could
not preserve the structural organization of the testicular tissue.
Collapse
Affiliation(s)
- Larissa Benvenutti
- Laboratório de Biotecnologia da Reprodução (LBR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, Santa Catarina, Brazil
| | - Rafael Alonso Salvador
- Laboratório de Biotecnologia da Reprodução (LBR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, Santa Catarina, Brazil
| | - David Til
- Laboratório de Biotecnologia da Reprodução (LBR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, Santa Catarina, Brazil
| | - Alfred Paul Senn
- Laboratório de Biotecnologia da Reprodução (LBR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, Santa Catarina, Brazil
| | - David Rivero Tames
- Laboratório de Biotecnologia da Reprodução (LBR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, Santa Catarina, Brazil
| | | | - Vera Lúcia Lângaro Amaral
- Laboratório de Biotecnologia da Reprodução (LBR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, Santa Catarina, Brazil
| |
Collapse
|
43
|
Devi L, Makala H, Pothana L, Nirmalkar K, Goel S. Comparative efficacies of six different media for cryopreservation of immature buffalo (Bubalus bubalis) calf testis. Reprod Fertil Dev 2018; 28:872-885. [PMID: 25482277 DOI: 10.1071/rd14171] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 10/20/2014] [Indexed: 12/20/2022] Open
Abstract
Buffalo calves have a high mortality rate (~80%) in commercial dairies and testis cryopreservation can provide a feasible option for the preservation of germplasm from immature males that die before attaining sexual maturity. The aim of the present study was to evaluate combinations of 10 or 20% dimethylsulfoxide (DMSO) with 0, 20 or 80% fetal bovine serum (FBS) for cryopreservation of immature buffalo testicular tissues, subjected to uncontrolled slow freezing. Tissues cryopreserved in 20% DMSO with 20% FBS (D20S20) showed total, tubular and interstitial cell viability, number of early apoptotic and DNA-damaged cells, surviving germ and proliferating cells and expression of testicular cell-specific proteins (POU class 5 homeobox (POU5F1), vimentin (VIM) and actin α2 (ACTA2)) similar to that of fresh cultured control (FCC; P>0.05). Expression of cytochrome P450, family 11, subfamily A (CYP11A1) protein and testosterone assay showed that only tissues cryopreserved in D20S20 had Leydig cells and secretory functions identical to that of FCC (P>0.05). High expression of superoxide dismutase2 (SOD2), cold-inducible RNA-binding protein (CIRBP) and RNA-binding motif protein3 (RBM3) proteins in cryopreserved tissues indicated involvement of cell signalling pathways regulating cellular protective mechanisms. Similarity in expression of pro-apoptosis proteins transcription factor tumour protein P53 (TP53) and BCL2-associated X protein (BAX) in D20S20 cryopreserved tissues to that of FCC (P>0.05) suggested lower apoptosis and DNA damage as key reasons for superior cryopreservation.
Collapse
Affiliation(s)
- Lalitha Devi
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Himesh Makala
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Lavanya Pothana
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Khemlal Nirmalkar
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Sandeep Goel
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| |
Collapse
|
44
|
Del Vento F, Vermeulen M, de Michele F, Giudice MG, Poels J, des Rieux A, Wyns C. Tissue Engineering to Improve Immature Testicular Tissue and Cell Transplantation Outcomes: One Step Closer to Fertility Restoration for Prepubertal Boys Exposed to Gonadotoxic Treatments. Int J Mol Sci 2018; 19:ijms19010286. [PMID: 29346308 PMCID: PMC5796232 DOI: 10.3390/ijms19010286] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 01/16/2018] [Accepted: 01/16/2018] [Indexed: 12/15/2022] Open
Abstract
Despite their important contribution to the cure of both oncological and benign diseases, gonadotoxic therapies present the risk of a severe impairment of fertility. Sperm cryopreservation is not an option to preserve prepubertal boys’ reproductive potential, as their seminiferous tubules only contain spermatogonial stem cells (as diploid precursors of spermatozoa). Cryobanking of human immature testicular tissue (ITT) prior to gonadotoxic therapies is an accepted practice. Evaluation of cryopreserved ITT using xenotransplantation in nude mice showed the survival of a limited proportion of spermatogonia and their ability to proliferate and initiate differentiation. However, complete spermatogenesis could not be achieved in the mouse model. Loss of germ cells after ITT grafting points to the need to optimize the transplantation technique. Tissue engineering, a new branch of science that aims at improving cellular environment using scaffolds and molecules administration, might be an approach for further progress. In this review, after summarizing the lessons learned from human prepubertal testicular germ cells or tissue xenotransplantation experiments, we will focus on the benefits that might be gathered using bioengineering techniques to enhance transplantation outcomes by optimizing early tissue graft revascularization, protecting cells from toxic insults linked to ischemic injury and exploring strategies to promote cellular differentiation.
Collapse
Affiliation(s)
- Federico Del Vento
- Gynecology-Andrology Unit, Medical School, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium; (F.D.V.); (M.V.); (F.d.M.); (M.G.G.)
| | - Maxime Vermeulen
- Gynecology-Andrology Unit, Medical School, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium; (F.D.V.); (M.V.); (F.d.M.); (M.G.G.)
| | - Francesca de Michele
- Gynecology-Andrology Unit, Medical School, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium; (F.D.V.); (M.V.); (F.d.M.); (M.G.G.)
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium;
| | - Maria Grazia Giudice
- Gynecology-Andrology Unit, Medical School, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium; (F.D.V.); (M.V.); (F.d.M.); (M.G.G.)
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium;
| | - Jonathan Poels
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium;
| | - Anne des Rieux
- Advanced Drug Delivery and Biomaterials Unit, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Christine Wyns
- Gynecology-Andrology Unit, Medical School, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium; (F.D.V.); (M.V.); (F.d.M.); (M.G.G.)
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium;
- Correspondence: ; Tel.: +32-2-764-95-01
| |
Collapse
|
45
|
Kaneko H, Kikuchi K, Nakai M, Fuchimoto D, Suzuki S, Sembon S, Noguchi J, Onishi A. Establishment of a strain of haemophilia-A pigs by xenografting of foetal testicular tissue from neonatally moribund cloned pigs. Sci Rep 2017; 7:17026. [PMID: 29208927 PMCID: PMC5717049 DOI: 10.1038/s41598-017-17017-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 11/20/2017] [Indexed: 12/18/2022] Open
Abstract
Grafting of testicular tissue into immunodeficient mice makes it possible to obtain functional sperm from immature donor animals that cannot be used for reproduction. We have developed a porcine model of human haemophilia A (haemophilia-A pigs) by nuclear transfer cloning from foetal fibroblasts after disruption of the X-linked coagulation factor VIII (F8) gene. Despite having a recessive condition, female F8+/- cloned pigs died of severe bleeding at an early age, as was the case for male F8-/Y cloned pigs, thus making it impossible to obtain progeny. In this study, therefore, we produced sperm from F8-/Y cloned pigs by grafting their foetal testicular tissue into nude mice. Two F8+/- female pigs were generated from oocytes injected with xenogeneic sperm. Unlike the F8+/- cloned pigs, they remained asymptomatic, and delivered five F8-/Y and four F8+/- pigs after being crossed with wild-type boars. The descendant F8-/Y pigs conserved the haemophilia phenotype. Thus, the present F8+/- pigs show resolution of the phenotypic abnormality, and will facilitate production of F8-/Y pigs as founders of a strain of haemophilia-A pigs for the development of new therapeutics for haemophilia A. This strategy will be applicable to other genetically modified pigs.
Collapse
Affiliation(s)
- Hiroyuki Kaneko
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, 305-8602, Japan.
| | - Kazuhiro Kikuchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, 305-8602, Japan.,The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi, 753-8515, Japan
| | - Michiko Nakai
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, 305-8602, Japan.,NARO, Tsukuba, Ibaraki, 305-8517, Japan
| | - Daiichiro Fuchimoto
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, 305-8602, Japan.
| | - Shunichi Suzuki
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, 305-8602, Japan.
| | - Shoichiro Sembon
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, 305-8602, Japan.,NARO, Tsukuba, Ibaraki, 305-8517, Japan
| | - Junko Noguchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, 305-8602, Japan
| | - Akira Onishi
- Nihon University, College of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
| |
Collapse
|
46
|
Update on fertility restoration from prepubertal spermatogonial stem cells: How far are we from clinical practice? Stem Cell Res 2017; 21:171-177. [DOI: 10.1016/j.scr.2017.01.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/09/2017] [Accepted: 01/23/2017] [Indexed: 01/15/2023] Open
|
47
|
Kaneko H, Kikuchi K, Men NT, Nakai M, Noguchi J, Kashiwazaki N, Ito J. Production of sperm from porcine fetal testicular tissue after cryopreservation and grafting into nude mice. Theriogenology 2017; 91:154-162. [DOI: 10.1016/j.theriogenology.2016.12.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/29/2016] [Accepted: 12/29/2016] [Indexed: 02/06/2023]
|
48
|
Vermeulen M, Poels J, de Michele F, des Rieux A, Wyns C. Restoring Fertility with Cryopreserved Prepubertal Testicular Tissue: Perspectives with Hydrogel Encapsulation, Nanotechnology, and Bioengineered Scaffolds. Ann Biomed Eng 2017; 45:1770-1781. [PMID: 28070774 DOI: 10.1007/s10439-017-1789-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/03/2017] [Indexed: 10/20/2022]
Abstract
New and improved oncological therapies are now able to cure more than 80% of cancer-affected children in Europe. However, such treatments are gonadotoxic and result in fertility issues, especially in boys who are not able to provide a sperm sample before starting chemo/radiotherapy because of their prepubertal state. For these boys, cryopreservation of immature testicular tissue (ITT) is the only available option, aiming to preserve spermatogonial stem cells (SSCs). Both slow-freezing and vitrification have been investigated to this end and are now applied in a clinical setting for SSC cryopreservation. Research now has to focus on methods that will allow fertility restoration. This review discusses different studies that have been conducted on ITT transplantation, including those using growth factor supplementation like free molecules, or tissue encapsulation with or without nanoparticles, as well as the possibility of developing a bioartificial testis that can be used for in vitro gamete production or in vivo transplantation.
Collapse
Affiliation(s)
- Maxime Vermeulen
- Gynecology-Andrology Research Unit, Medical School, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Jonathan Poels
- Gynecology-Andrology Research Unit, Medical School, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200, Brussels, Belgium.,Department of Gynecology-Andrology, Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium
| | - Francesca de Michele
- Gynecology-Andrology Research Unit, Medical School, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200, Brussels, Belgium.,Department of Gynecology-Andrology, Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium
| | - Anne des Rieux
- Advanced Drug Delivery and Biomaterials Unit, Louvain Drug Research Institute, Université Catholique de Louvain, 1200, Brussels, Belgium.,Institute of Condensed Matter and Nanosciences, Université Catholique de Louvain, 1348, Louvain-la-Neuve, Belgium
| | - Christine Wyns
- Gynecology-Andrology Research Unit, Medical School, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200, Brussels, Belgium. .,Department of Gynecology-Andrology, Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium.
| |
Collapse
|
49
|
Devi L, Goel S. Fertility preservation through gonadal cryopreservation. Reprod Med Biol 2016; 15:235-251. [PMID: 29259441 PMCID: PMC5715865 DOI: 10.1007/s12522-016-0240-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/01/2016] [Indexed: 12/20/2022] Open
Abstract
Fertility preservation is an area of immense interest in today's society. The most effective and established means of fertility preservation is cryopreservation of gametes (sperm and oocytes) and embryos. Gonadal cryopreservation is yet another means for fertility preservation, especially if the gonadal function is threatened by premature menopause, gonadotoxic cancer treatment, surgical castration, or diseases. It can also aid in the preservation of germplasm of animals that die before attaining sexual maturity. This is especially of significance for valuable, rare, and endangered animals whose population is affected by high neonatal/juvenile mortality because of diseases, poor management practices, or inbreeding depression. Establishing genome resource banks to conserve the genetic status of wild animals will provide a critical interface between ex-situ and in-situ conservation strategies. Cryopreservation of gonads effectively lengthens the genetic lifespan of individuals in a breeding program even after their death and contributes towards germplasm conservation of prized animals. Although the studies on domestic animals are quite promising, there are limitations for developing cryopreservation strategies in wild animals. In this review, we discuss different options for gonadal tissue cryopreservation with respect to humans and to laboratory, domestic, and wild animals. This review also covers recent developments in gonadal tissue cryopreservation and transplantation, providing a systematic view and the advances in the field with the possibility for its application in fertility preservation and for the conservation of germplasm in domestic and wild species.
Collapse
Affiliation(s)
- Lalitha Devi
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular BiologyCouncil for Scientific and Industrial ResearchUppal Road500 007HyderabadIndia
| | - Sandeep Goel
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular BiologyCouncil for Scientific and Industrial ResearchUppal Road500 007HyderabadIndia
| |
Collapse
|
50
|
Pothana L, Devi L, Venna NK, Pentakota N, Varma VP, Jose J, Goel S. Replacement of serum with ocular fluid for cryopreservation of immature testes. Cryobiology 2016; 73:356-366. [PMID: 27693391 DOI: 10.1016/j.cryobiol.2016.09.169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/26/2016] [Accepted: 09/26/2016] [Indexed: 11/25/2022]
Abstract
Cryopreservation of immature testis is a feasible approach for germplasm preservation of male animals. Combinations of dimethyl sulfoxide (DMSO) and foetal bovine serum (FBS) are used for testis cryopreservation. However, an alternative to FBS is needed, because FBS is expensive. Buffalo ocular fluid (BuOF), a slaughter house by-product, could be an economical option. The objective of the present study was to assess whether BuOF can replace FBS for cryopreservation of immature mouse (Mus musculus), rat (Rattus norvegicus), and buffalo (Bubalus bubalis) testes. Results showed that rodent and buffalo testes frozen in DMSO (10% for rodents and 20% for buffalo) with 20% FBS or BuOF had similar numbers of viable and DNA-damaged cells (P > 0.05). The expression of cell proliferation- (PCNA) and apoptosis-specific proteins (Annexin V and BAX/BCL2 ratio) were also comparable in mouse and buffalo testes frozen in DMSO with FBS or BuOF (P > 0.05). Interestingly, rat testis frozen in DMSO with BuOF had lower expression of Annexin V protein than testis frozen in DMSO with FBS (P < 0.05). The percentage of meiotic germ cells (pachytene-stage spermatocytes) in xenografts from testis frozen either in DMSO with BuOF or FBS did not significantly differ in rats or buffalo (P > 0.05). These findings provide evidence that BuOF has potential to replace FBS for cryopreservation of immature rodent and buffalo testis. Further investigation is needed to explore whether BuOF can replace FBS for testis cryopreservation of other species.
Collapse
Affiliation(s)
- Lavanya Pothana
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Lalitha Devi
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Naresh Kumar Venna
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Niharika Pentakota
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Vivek Phani Varma
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Jedy Jose
- Animal House, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Sandeep Goel
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India.
| |
Collapse
|