Extracellular matrix and extracellular matrix-derived materials in reproductive medicine | Placenta and Reproductive Medicine

Extracellular matrix and extracellular matrix-derived materials in reproductive medicine

Review of ECM in reproductive medicine

Authors

  • Zhengyan Hu The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
  • Rui Gao The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
  • Hanxiao Chen The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
  • Minqi Chen The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
  • Lang Qin The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China

DOI:

https://doi.org/10.54844/prm.2022.0142

Keywords:

extracellular matrix, reproductive medicine, tissue engineering

Abstract

Infertility has become a worldwide issue, and many patients cannot benefit from assisted reproductive technologies (ART). The extracellular matrix (ECM) is critical in tissue organization and remodeling. The female reproductive system plays an important role in menstruation, pregnancy, and ovulation and may influence fertility. In addition, ECM has a wide variety of components, good biological properties, and extensive application experience as a biomaterial. In-depth research on the ECM in the female reproductive system and the development and application of ECM-derived materials may provide new ideas for solving infertility problems. This review aimed to summarize the regulation and changes of ECM in the uterus and ovary, and to discuss the progress of research on ECM-derived materials in reproductive tissue engineering. An extensive search in PubMed and Embase was conducted using keywords including extracellular matrix, uterus, ovary, tissue engineering, and material. We are devoted to combining research on ECM-derived materials with clinical practice and intend to provide ideas for solving clinical problems in reproductive medicine.

References

Vander Borght M, Wyns C. Fertility and infertility: definition and epidemiology. Clin Biochem. 2018;62:2–10.

Steptoe PC, Edwards RG. Birth after the reimplantation of a human embryo. Lancet. 1978;2(8085):366.

Palermo G, Joris H, Devroey P, van Steirteghem AC. Pregnancies after intracytoplasmic injection of single spermatozoon into an oocyte. Lancet. 1992;340(8810):17–18.

Karamanos NK, Theocharis AD, Piperigkou Z, et al. A guide to the composition and functions of the extracellular matrix. FEBS J. 2021;288(24):6850–6912.

O’Connor BB, Pope BD, Peters MM, Ris-Stalpers C, Parker KK. The role of extracellular matrix in normal and pathological pregnancy: future applications of microphysiological systems in reproductive medicine. Exp Biol Med (Maywood). 2020;245(13):1163–1174.

Chen C. Pregnancy after human oocyte cryopreservation. Lancet. 1986;1(8486):884–886.

Bedaiwy MA, Shahin AY, Falcone T. Reproductive organ transplantation: advances and controversies. Fertil Steril. 2008;90(6):2031–2055.

Badylak SF, Freytes DO, Gilbert TW. Extracellular matrix as a biological scaffold material: structure and function. Acta Biomater. 2009;5(1):1–13.

Gelse K, Pöschl E, Aigner T. Collagens—structure, function, and biosynthesis. Adv Drug Deliv Rev. 2003;55(12):1531–1546.

Vasvani S, Kulkarni P, Rawtani D. Hyaluronic acid: a review on its biology, aspects of drug delivery, route of administrations and a special emphasis on its approved marketed products and recent clinical studies. Int J Biol Macromol. 2020;151:1012–1029.

Wen Q, Mithieux SM, Weiss AS. Elastin biomaterials in dermal repair. Trends Biotechnol. 2020;38(3):280–291.

Vindin H, Mithieux SM, Weiss AS. Elastin architecture. Matrix Biol. 2019;84:4–16.

Almine JF, Bax DV, Mithieux SM, et al. Elastin-based materials. Chem Soc Rev. 2010;39(9):3371–3379.

Aumailley M. The laminin family. Cell Adh Migr. 2013;7(1):48–55.

Yurchenco PD, McKee KK, Reinhard JR, Rüegg MA. Laminin-deficient muscular dystrophy: molecular pathogenesis and structural repair strategies. Matrix Biol. 2018;71–72:174–187.

Patten J, Wang K. Fibronectin in development and wound healing. Adv Drug Deliv Rev. 2021;170:353–368.

Werb Z. ECM and cell surface proteolysis: regulating cellular ecology. Cell. 1997;91(4):439–442.

Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ Res. 2003;92(8):827–839.

Edwards DR, Handsley MM, Pennington CJ. The ADAM metalloproteinases. Mol Aspects Med. 2008;29(5):258–289.

Mead TJ, Apte SS. ADAMTS proteins in human disorders. Matrix Biol. 2018;71–72:225–239.

Gross J, Lapiere CM. Collagenolytic activity in amphibian tissues: a tissue culture assay. Proc Natl Acad Sci U S A. 1962;48(6):1014–1022.

Cui N, Hu M, Khalil RA. Biochemical and biological attributes of matrix metalloproteinases. Prog Mol Biol Transl Sci. 2017;147:1–73.

Zitka O, Kukacka J, Krizkova S, et al. Matrix metalloproteinases. Curr Med Chem. 2010;17(31):3751–3768.

Arpino V, Brock M, Gill SE. The role of TIMPs in regulation of extracellular matrix proteolysis. Matrix Biol. 2015;44–46:247–254.

Ågren MS, Schnabel R, Christensen LH, Mirastschijski U. Tumor necrosis factor-α-accelerated degradation of type I collagen in human skin is associated with elevated matrix metalloproteinase (MMP)-1 and MMP-3 ex vivo. Eur J Cell Biol. 2015;94(1):12–21.

Mirastschijski U, Lupše B, Maedler K, et al. Matrix metalloproteinase-3 is key effector of TNF-α-induced collagen degradation in skin. Int J Mol Sci. 2019;20(20):5234.

Li X, Guo L, Yang X, et al. TGF-β1-induced connexin43 promotes scar formation via the Erk/MMP-1/collagen III pathway. J Oral Rehabil. 2020;47(Suppl 1):99–106.

Chen LC, Shibu MA, Liu CJ, et al. ERK1/2 mediates the lipopolysaccharideinduced upregulation of FGF-2, uPA, MMP-2, MMP-9 and cellular migration in cardiac fibroblasts. Chem Biol Interact. 2019;306:62–69.

Salamonsen LA, Zhang J, Hampton A, Lathbury L. Regulation of matrix metalloproteinases in human endometrium. Hum Reprod. 2000;15(Suppl3):112–119.

Salamonsen LA, Woolley DE. Matrix metalloproteinases in normal menstruation. Hum Reprod. 1996;11(Suppl 2):124–133.

Obokata A, Watanabe J, Nishimura Y, Arai T, Kawaguchi M, Kuramoto H. Significance of matrix metalloproteinase-7[correction of matrix metalloproteinase-2],-11 and tissue inhibitor of metalloproteinase-1 expression in normal, hyperplastic and neoplastic endometrium. Anticancer Res. 2007;27(1A):95–105.

Chakraborti S, Mandal M, Das S, Mandal A, Chakraborti T. Regulation of matrix metalloproteinases: an overview. Mol Cell Biochem. 2003;253(1):269–285.

Yoshii N, Hamatani T, Inagaki N, et al. Successful implantation after reducing matrix metalloproteinase activity in the uterine cavity. Reprod Biol Endocrinol. 2013;11:37.

Basu J, Agamasu E, Bendek B, et al. Correlation Between Placental Matrix Metalloproteinase 9 and Tumor Necrosis Factor-α Protein Expression Throughout Gestation in Normal Human Pregnancy. Reprod Sci. 2018;25(4):621–627.

Xu P, Alfaidy N, Challis JR. Expression of matrix metalloproteinase (MMP)-2 and MMP-9 in human placenta and fetal membranes in relation to preterm and term labor. J Clin Endocrinol Metab. 2002;87(3):1353–1361.

Wang W, Vilella F, Alama P, et al. Single-cell transcriptomic atlas of the human endometrium during the menstrual cycle. Nat Med. 2020;26(10):1644–1653.

Sternberg AK, Buck VU, Classen-Linke I, Leube RE. How mechanical forces change the human endometrium during the menstrual cycle in preparation for embryo implantation. Cells. 2021;10(8):2008.

Schatz F, Guzeloglu-Kayisli O, Arlier S, Kayisli UA, Lockwood CJ. The role of decidual cells in uterine hemostasis, menstruation, inflammation, adverse pregnancy outcomes and abnormal uterine bleeding. Hum Reprod Update. 2016;22(4):497–515.

Burrows TD, King A, Smith SK, Loke YW. Human trophoblast adhesion to matrix proteins: inhibition and signal transduction. Hum Reprod. 1995;10(9):2489–2500.

Sinai Talaulikar V, Kronenberger K, Bax BE, Moss R, Manyonda I. Differences in collagen ultrastructure of human first trimester decidua basalis and parietalis: implications for trophoblastic invasion of the placental bed. J Obstet Gynaecol Res. 2014;40(1):80–88.

Tanaka T, Wang C, Umesaki N. Remodeling of the human endometrial epithelium is regulated by laminin and type IV collagen. Int J Mol Med. 2009;23(2):173–180.

Yang ZS, Pan HY, Shi WW, et al. Regulation and function of laminin A5 during mouse and human decidualization. Int J Mol Sci. 2021;23(1):199.

Sanders B. Uterine factors and infertility. J Reprod Med. 2006;51(3):169–176.

Liu KE, Hartman M, Hartman A. Management of thin endometrium in assisted reproduction: a clinical practice guideline from the Canadian Fertility and Andrology Society. Reprod Biomed Online. 2019;39(1):49–62.

Berman JM. Intrauterine adhesions. Semin Reprod Med. 2008;26(4):349–355.

Nejat EJ, Zapantis G, Rybak EA, Meier UT. It’s time to pay attention to the endometrium, including the nucleolar channel system. Fertil Steril. 2011;96(6):e165;authorreplye166.

Jiang P, Tang X, Wang H, et al. Collagen-binding basic fibroblast growth factor improves functional remodeling of scar red endometrium in uterine infertile women: a pilot study. Sci China Life Sci. 2019;62(12):1617–1629.

Cimadomo D, Craciunas L, Vermeulen N, Vomstein K, Toth B. Definition, diagnostic and therapeutic options in recurrent implantation failure: an international survey of clinicians and embryologists. Hum Reprod. 2021;36(2):305–317.

Turpeenniemi-Hujanen T, Feinberg RF, Kauppila A, Puistola U. Extracellular matrix interactions in early human embryos: implications for normal implantation events. Fertil Steril. 1995;64(1):132–138.

Fouladi-Nashta AA, Raheem KA, Marei WF, Ghafari F, Hartshorne GM. Regulation and roles of the hyaluronan system in mammalian reproduction. Reproduction. 2017;153(2):R43–R58.

Zhang C, Duan E, Cao Y, Jiang G, Zeng G. Effect of 32/67 kDa laminin-binding protein antibody on mouse embryo implantation. J Reprod Fertil. 2000;119(1):137–142.

Klemmt PA, Carver JG, Koninckx P, McVeigh EJ, Mardon HJ. Endometrial cells from women with endometriosis have increased adhesion and proliferative capacity in response to extracellular matrix components: towards a mechanistic model for endometriosis progression. Hum Reprod. 2007;22(12):3139–3147.

Dimitriadis E, Menkhorst E, Saito S, Kutteh WH, Brosens JJ. Recurrent pregnancy loss. Nat Rev Dis Primers. 2020;6(1):98.

Jerzak M, Rechberger T, Górski A. Intravenous immunoglobulin therapy influences T cell adhesion to extracellular matrix in women with a history of recurrent spontaneous abortions. Am J Reprod Immunol. 2000;44(6):336–341.

Ozawa F, Mori R, Obayashi Y, et al. Clinical significance of MMPs (matrix metalloproteinases) and TIMPs (tissue inhibitors of metalloproteinases) with recurrent miscarriage. Nagoya: The 71st Annual Congress of the Japan Society of Obstetrics and Gynecology, 2019.

Endo T, Kiya T, Goto T, et al. Significance of matrix metalloproteinases in the pathophysiology of the ovary and uterus. Reprod Med Biol. 2006;5(4):235–243.

Vos MC, van der Wurff AA, Last JT, et al. Immunohistochemical expression of MMP-14 and MMP-2, and MMP-2 activity during human ovarian follicular development. Reprod Biol Endocrinol. 2014;12:12.

Riley SC, Thomassen R, Bae SE, Leask R, Pedersen HG, Watson ED. Matrix metalloproteinase-2 and-9 secretion by the equine ovary during follicular growth and prior to ovulation. Anim Reprod Sci. 2004;81(3–4):329–339.

McCord L, Park E, Jo M, Brannstrom M, Curry TE. Expression of matrix metalloproteinase 10 (Mmp10) during the periovulatory period. Biol Reprod. 2010;83(Suppl_1):642.

Amargant F, Manuel SL, Tu Q, et al. Ovarian stiffness increases with age in the mammalian ovary and depends on collagen and hyaluronan matrices. Aging Cell. 2020;19(11):e13259.

Irving-Rodgers HF, Rodgers RJ. Extracellular matrix in ovarian follicular development and disease. Cell Tissue Res. 2005;322(1):89–98.

Irving-Rodgers HF, Mussard ML, Kinder JE, Rodgers RJ. Composition and morphology of the follicular basal lamina during atresia of bovine antral follicles. Reproduction. 2002;123(1):97–106.

Jankowska K. Premature ovarian failure. Menopause Rev. 2017;16(2):51–56.

Zhou F, Shi LB, Zhang SY. Ovarian fibrosis: a phenomenon of concern. Chin Med J (Engl). 2017;130(3):365–371.

Patel S. Polycystic ovary syndrome (PCOS), an inflammatory, systemic, lifestyle endocrinopathy. J Steroid Biochem Mol Biol. 2018;182:27–36.

Lewandowski KC, Komorowski J, O’Callaghan CJ, et al. Increased circulating levels of matrix metalloproteinase-2 and-9 in women with the polycystic ovary syndrome. J Clin Endocrinol Metab. 2006;91(3):1173–1177.

Xu L, Ding L, Wang L, et al. Umbilical cord-derived mesenchymal stem cells on scaffolds facilitate collagen degradation via upregulation of MMP-9 in rat uterine scars. Stem Cell Res Ther. 2017;8(1):84.

Cao Y, Sun H, Zhu H, et al. Allogeneic cell therapy using umbilical cord MSCs on collagen scaffolds for patients with recurrent uterine adhesion: a phase I clinical trial. Stem Cell Res Ther. 2018;9(1):192.

Zhang Y, Shi L, Lin X, et al. Unresponsive thin endometrium caused by Asherman syndrome treated with umbilical cord mesenchymal stem cells on collagen scaffolds: a pilot study. Stem Cell Res Ther. 2021;12(1):420.

Oktem O, Oktay K. The role of extracellular matrix and activin-A in in vitro growth and survival of murine preantral follicles. Reprod Sci. 2007;14(4):358–366.

Heymann D, Vidal L, Or Y, Shoham Z. Hyaluronic acid in embryo transfer media for assisted reproductive technologies. Cochrane Database Syst Rev. 2020;9(9):CD007421.

Shakouri-Motlagh A, O’Connor AJ, Brennecke SP, Kalionis B, Heath DE. Native and solubilized decellularized extracellular matrix: a critical assessment of their potential for improving the expansion of mesenchymal stem cells. Acta Biomater. 2017;55:1–12.

Miyazaki K, Maruyama T. Partial regeneration and reconstruction of the rat uterus through recellularization of a decellularized uterine matrix. Biomaterials. 2014;35(31):8791–8800.

Miki F, Maruyama T, Miyazaki K, et al. The orientation of a decellularized uterine scaffold determines the tissue topology and architecture of the regenerated uterus in rats. Biol Reprod. 2019;100(5):1215–1227.

Padma AM, Tiemann TT, Alshaikh AB, Akouri R, Song MJ, Hellström M. Protocols for rat uterus isolation and decellularization: applications for uterus tissue engineering and 3D cell culturing. Methods Mol Biol. 2018;1577:161–175.

Pors SE, Ramløse M, Nikiforov D, et al. Initial steps in reconstruction of the human ovary: survival of pre-antral stage follicles in a decellularized human ovarian scaffold. Hum Reprod. 2019;34(8):1523–1535.

Oktay K, Bedoschi G, Pacheco F, Turan V, Emirdar V. First pregnancies, live birth, and in vitro fertilization outcomes after transplantation of frozen-banked ovarian tissue with a human extracellular matrix scaffold using robot-assisted minimally invasive surgery. Am J Obstet Gynecol. 2016;214(1):94.e1–94.e9.

Vanacker J, Luyckx V, Dolmans MM, et al. Transplantation of an alginate-matrigel matrix containing isolated ovarian cells: first step in developing a biodegradable scaffold to transplant isolated preantral follicles and ovarian cells. Biomaterials. 2012;33(26):6079–6085.

Choi JK, Agarwal P, Huang H, Zhao S, He X. The crucial role of mechanical heterogeneity in regulating follicle development and ovulation with engineered ovarian microtissue. Biomaterials. 2014;35(19):5122–5128.

Jamalzaei P, Rezazadeh Valojerdi M, Montazeri L, Baharvand H. Applicability of hyaluronic acid-alginate hydrogel and ovarian cells for in vitro development of mouse preantral follicles. Cell J. 2020;22(Suppl 1):49–60.

Arora B, Tandon R, Attri P, Bhatia R. Chemical crosslinking: role in protein and peptide science. Curr Protein Pept Sci. 2017;18(9):946–955.

Koide T, Daito M. Effects of various collagen crosslinking techniques on mechanical properties of collagen film. Dent Mater J. 1997;16(1):1–9.

Reddy N, Reddy R, Jiang Q. Crosslinking biopolymers for biomedical applications. Trends Biotechnol. 2015;33(6):362–369.

Jiang Q, Reddy N, Yang Y. Cytocompatible cross-linking of electrospun zein fibers for the development of water-stable tissue engineering scaffolds. Acta Biomater. 2010;6(10):4042–4051.

Delgado LM, Bayon Y, Pandit A, Zeugolis DI. To cross-link or not to cross-link? Cross-linking associated foreign body response of collagenbased devices. Tissue Eng Part B Rev. 2015;21(3):298–313.

Kobayashi T, Chanmee T, Itano N. Hyaluronan: metabolism and function. Biomolecules. 2020;10(11):1525.

Segura T, Anderson BC, Chung PH, Webber RE, Shull KR, Shea LD. Crosslinked hyaluronic acid hydrogels: a strategy to functionalize and pattern. Biomaterials. 2005;26(4):359–371.

Lou J, Stowers R, Nam S, Xia Y, Chaudhuri O. Stress relaxing hyaluronic acid-collagen hydrogels promote cell spreading, fiber remodeling, and focal

adhesion formation in 3D cell culture. Biomaterials. 2018;154:213–222.

Ge F, Lu Y, Li Q, Zhang X. Decellularized extracellular matrices for tissue engineering and regeneration. Adv Exp Med Biol. 2020;1250:15–31.

Liu C, Pei M, Li Q, Zhang Y. Decellularized extracellular matrix mediates tissue construction and regeneration. Front Med. 2022;16(1):56–82.

Ma B, Wang X, Wu C, Chang J. Crosslinking strategies for preparation of extracellular matrix-derived cardiovascular scaffolds. Regen Biomater. 2014;1(1):81–89.

Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8(9):726–736.

Talovic M, Patel K, Schwartz M, Madsen J, Garg K. Decellularized extracellular matrix gelloids support mesenchymal stem cell growth and function in vitro. J Tissue Eng Regen Med. 2019;13(10):1830–1842.

Lai Y, Sun Y, Skinner CM, et al. Reconstitution of marrow-derived extracellular matrix ex vivo: a robust culture system for expanding largescale highly functional human mesenchymal stem cells. Stem Cells Dev. 2010;19(7):1095–1107.

Lin H, Yang G, Tan J, Tuan RS. Influence of decellularized matrix derived from human mesenchymal stem cells on their proliferation, migration and multi-lineage differentiation potential. Biomaterials. 2012;33(18):4480–4489.

Dunn A, Talovic M, Patel K, Patel A, Marcinczyk M, Garg K. Biomaterial and stem cell-based strategies for skeletal muscle regeneration. J Orthop Res. 2019;37(6):1246–1262.

Caliari SR, Burdick JA. A practical guide to hydrogels for cell culture. Nat Methods. 2016;13(5):405–414.

Cao H, Duan L, Zhang Y, Cao J, Zhang K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther. 2021;6(1):426.

Huang Q, Zou Y, Arno MC, et al. Hydrogel scaffolds for differentiation of adipose-derived stem cells. Chem Soc Rev. 2017;46(20):6255–6275.

Lv H, Wu B, Song J, Wu W, Cai W, Xu J. Hydrogel, a novel therapeutic and delivery strategy, in the treatment of intrauterine adhesions. J Mater Chem B. 2021;9(33):6536–6552.

Chan M, Keane T, Jones B, Smith JR, Stevens M, Saso S. Bioengineering approaches for female fertility preservation and infertility treatment. BJOG. 2019;126(S2):196.

Nikukar H. Regenerative medicine for female reproductive system. Accessed January 12, 2023. http://ijrm.ir/browse.php?mag_id=163&slc_lang=en&sid=1

Yoshimasa Y, Maruyama T. Bioengineering of the uterus. Reprod Sci. 2021;28(6):1596–1611.

Xin L, Lin X, Pan Y, et al. A collagen scaffold loaded with human umbilical cord-derived mesenchymal stem cells facilitates endometrial regeneration and restores fertility. Acta Biomater. 2019;92:160–171.

Donnez J, Dolmans MM. Fertility Preservation in Women. N Engl J Med. 2017;377(17):1657–1665.

Published

2023-01-31

How to Cite

1.
Hu Z, Gao R, Chen H, Chen M, Qin L. Extracellular matrix and extracellular matrix-derived materials in reproductive medicine: Review of ECM in reproductive medicine. PRM. 2023;2. doi:10.54844/prm.2022.0142

Issue

Section

Review Article

Downloads

Download data is not yet available.
REVIEW ARTICLE

Extracellular matrix and extracellular matrix-derived materials in reproductive medicine


Zhengyan Hu1,2,3, Rui Gao1,2,3, Hanxiao Chen1,2,3, Minqi Chen1,2,3, Lang Qin1,2

1The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China;

2Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, Sichuan Province, China;

3West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China


Address for correspondence:

Prof. Lang Qin, E-mail: cacier@163.com; https://orcid.org/0000-0003-1794-6700


Received: 28 August 2022

Accepted: 23 November 2022

Published: 31 January 2023


Abstract

Infertility has become a worldwide issue, and many patients cannot benefit from assisted reproductive technologies (ART). The extracellular matrix (ECM) is critical in tissue organization and remodeling. The female reproductive system plays an important role in menstruation, pregnancy, and ovulation and may influence fertility. In addition, ECM has a wide variety of components, good biological properties, and extensive application experience as a biomaterial. In-depth research on the ECM in the female reproductive system and the development and application of ECM-derived materials may provide new ideas for solving infertility problems. This review aimed to summarize the regulation and changes of ECM in the uterus and ovary, and to discuss the progress of research on ECM-derived materials in reproductive tissue engineering. An extensive search in PubMed and Embase was conducted using keywords including extracellular matrix, uterus, ovary, tissue engineering, and material. We are devoted to combining research on ECM-derived materials with clinical practice and intend to provide ideas for solving clinical problems in reproductive medicine.

Key words: extracellular matrix, reproductive medicine, tissue engineering

INTRODUCTION

With 8%–12% of couples of reproductive age worldwide currently suffering from infertility,[1] treatment of infertility has become a real challenge. In the past few decades, reproductive medicine has developed rapidly, with repeated breakthroughs in research in assisted reproductive technologies (ART).[2,3] However, patients with uterine infertility may not benefit from ART, which remains difficult to solve. Patients with damaged ovaries also cannot regain their fertility through ART alone.

Recently, the physiological role played by the extracellular matrix (ECM) in the process of implantation and pregnancy has been increasingly evidenced. ECM is a 3-dimensional (3D) polymeric network structure widely distributed in the body, providing structural and functional support to cells and tissues. It is critical in regulating cell processes, including proliferation, survival, differentiation, migration, angiogenesis, and immune response.[4] In the reproductive system, the ECM is dynamically regulated during the menstrual cycle, peri-implantation, pregnancy, and delivery, and its dysregulation is related to various diseases.[5] Therefore, research focused on ECM in the female reproductive system and infertility is becoming more prevalent.

Moreover, ECM provides another way of thinking about the treatment of infertility. Patients suffering uterine infertility or ovarian damage may benefit from other technologies, such as oocyte cryopreservation[6] and organ transplantation.[7] The further development of these technologies is closely related to tissue engineering and biomaterial science. Biological scaffolds derived from ECM have been shown to promote tissue repair and reconstruction in animal experiments and clinical studies. Due to its biodegradability and biocompatibility, ECM has already been applicated in tissue engineering and regenerative medicine.[8]

The components of ECM include collagen, proteoglycans (PGs) or glycosaminoglycans (GAGs), elastin, elastic fibers, laminin, and fibronectin.[4] Collagen has the mechanical function of providing many critical biomechanical properties in the body and the biological function to help tissue anchor and adhere.[9] Collagen has been applicated in biomaterial science and clinical medicine for years as an important natural renewable resource. Hyaluronic acid (HA) is an important type of GAG in ECM which has special functions in embryogenesis and tissue repair[10] and is a biomaterial with promising applications. Elastin is an insoluble protein component of ECM conferring elasticity to organs, whose mechanical properties may regulate cell properties, implying a positive effect on cell growth. It is the major constituent of elastic fibers, whose alignment influences cell phenotype, adhesion, and proliferation.[11,12] The application of elastin-based materials also has been reported.[13] Laminins are essential glycoproteins in the specialized ECM, basement membrane. They play a role in attaching ECM to the cell surface and serve as a platform for other ECM components to stably attach to, during which they regulate multiple cellular activities and signaling pathways.[14,15] Fibronectin is a protein that controls cell adhesion, spreading, migration, proliferation, and apoptosis.[16]

In summary, ECM is made up of a wide variety of components, has good biological properties and application experience. Furthermore, its regulation and physiological roles within the reproductive system are increasingly studied. Therefore, ECM may provide new ideas for developing reproductive medicine and facilitating material science research in reproductive medicine. This review aims to summarize the regulation and changes of ECM in the uterus and ovary, and to discuss the progress of research on ECM-derived materials in reproductive tissue engineering. We are devoted to combining research on ECM-derived materials with clinical practice, intending to provide ideas for solving clinical problems.

METHODOLOGY

An extensive literature search in PubMed and Embase was conducted using the keywords and their permutations shown in Table 1. Original articles and reviews in English were scanned, and conference proceedings were checked through Embase.

Table 1: Keywords used for literature searching
Section Keywords
Full text extracellular matrix; collagen; fibronectin; laminin; elastin; hyaluronic acid
Roles of ECM uterus; endometrium; ovary; pregnancy; ovulation
ECM-derived materials material
Application tissue engineering; ovary; uterus
ECM: extracellular matrix.

ROLES OF THE ECM IN THE FEMALE REPRODUCTIVE SYSTEM

Metabolism of ECM

Proteolysis of ECM components during physiological and pathological processes is accomplished by the assembly, structural remodeling, correction of overexpression, and release of bioactive fragments and growth factors.[17] In the process of ECM degradation, matrix metalloproteinases (MMPs), a disintegrin and metalloproteinase (ADAM), and a disintegrin and metalloprotease with thrombospondin motifs (ADAMTS) play important roles,[18,19,20] all of which belong to the superfamily of metzincin proteases, a family of multidomain zinc-dependent endo-proteases. Among them, the main enzymes to degrade ECM are MMPs,[21] which can degrade most of the important protein components of ECM, including collagen, fibronectin, elastin, and laminin.[22,23]

MMPs, ADAM and ADAMTS are regulated to maintain the homeostasis of ECM. Endogenous tissue inhibitors of matrix metalloproteinases (TIMPs) can selectively inhibit different subtypes of MMPs, ADAMs, and ADAMTS, therefore playing a role in matrix accumulation and proteolysis.[24] Multiple factors and pathways tightly regulate the expression level and activity of MMPs/TIMPs in vivo, and the MMPs/TIMPs balance will further affect ECM. For instance, tumor necrosis factor (TNF)-α may enhance the collagenolytic activity of MMP-1 via up-regulating the expression of MMP-3, and down-regulate the expression of TIMP-1 to degenerate collagen.[25,26] When the extracellular-signal-regulated protein kinase 1/2 (ERK1/2) pathway is blocked, the expression of MMP-1 is decreased, and collagen III accumulation is promoted,[27] while ERK1/2 may also mediate up-regulation of MMP-2 and MMP-9.[28] In the female reproductive system, the composition of ECM is similar but with some tissue specificity. The regulation of MMPs also varies due to distinct environments.

ECM in the uterus

Regulation of ECM in the uterus

In the endometrium, MMPs are secreted by endometrial cells. In physiological processes such as menstruation, endometrial remodeling, decidualization, and embryo implantation, MMPs play a significant role and are regulated by various factors.[29] MMP-1, -3, and -9 were specifically related to tissue degradation in menstrual endometrium and activated by interleukin-1 (IL-1) and TNF-α.[30] MMP-7, MMP-11, and TIMP-1 are related to the remodeling of the endometrium in the normal menstrual cycle.[31] MMPs have been proven to be regulated by sex steroid hormones during the menstrual cycle. For example, progesterone can inhibit the secretion and release of MMP-11 and -14. Estrogen can enhance this process via the up-regulation of progesterone receptors.[32] Besides, MMP-2 and MMP-9 are indispensable for embryo implantation. They are the key rate-limiting enzymes in ECM remodeling during implantation, while correction of the overactivity of MMP-2 and MMP-9 is associated with a higher rate of successful implantation.[33]

In the placenta, MMP-2 and MMP-9 are expressed in a cell-specific manner. It has been reported that a significant correlation between TNF-α and MMP-9 in the second trimester may contribute to a successful pregnancy outcome.[34] Increased expression of MMP-9 contributes to ECM degradation, thus promoting rupture of fetal membrane and separation of the placenta during delivery.[35]

Changes of ECM during pregnancy

Decidualization occurs during the implantation window, and stromal fibroblast-like cells differentiate into epithelial-like decidua cells.[36] To better receive the embryo, the endometrial epithelial cells lose polarity, and the intercellular space becomes wider and is inserted by the ECM.[37] The ECM composition has undergone overall changes, especially collagen type IV, laminin, and fibronectin.[37,38] According to staining results, collagen type IV appears earlier around the spiral arteries of the decidualized sheath and may be involved in arterial remodeling.[39] For successful trophoblast invasion, the arrangement of collagens in the decidua is uneven, with collagen fibrils thicker around the trophoblast.[40] Laminin undergoes a transient reduction in the secretory phase but increases to an abundant level during decidualization and after the invasion. As well as collagen IV, laminin may play an essential role in decidualization and invasion and has a remarkable effect on the enhancement of endometrial ECM stiffness.[37,41,42] Fibronectin can be secreted by decidual stromal cells and trophoblasts and is regarded to mediate the migration of trophoblasts.[39]

Pathological states in the uterus

The causes of uterine infertility include intrauterine adhesions, thin endometrium, uterine septa, uterine myomas, and endometrial polyps.[43,44] Among others, intrauterine adhesions and thin endometrium are usually related to damage to the endometrium caused by traumatic uterine cavity procedures.[45,46] When the endometrium is injured, the native cells are replaced by fibroblasts and myofibroblasts and produce a large amount of ECM, including collagen, leading to the formation of uterine scar, which may cause infertility.[47]

Recurrent implantation failure (RIF) is a definition that applies to women undergoing ART. It is suggested that a case of RIF can be diagnosed when > 3 failed embryo transfers with high-quality embryos or the failed transfers of ≥ 10 embryos in multiple transfers happen.[48] RIF due to endometrial factors has been a challenge in reproductive medicine. Some studies have suggested that dysregulation of the endometrial ECM may limit trophoblast invasion, leading to RIF.[49] HA may have a two-sided effect on pregnancy and play a role as a sticky matrix for embryos to attach, therefore reducing implantation failure. However, the accumulation of HA may be related to early embryo loss.[50] Laminin, fibronectin, integrin, and other ECM components may influence embryo implantation directly or indirectly.[51,52]

Recurrent pregnancy loss (RPL) is the failure of two or more clinically recognized pregnancies.[53] The exact mechanism of ECM involvement in RPL is unclear. However, studies have shown that the increased adhesion between ECM and T cells may be part of the immunological mechanism of RPL.[54] Besides, the staining of MMP-2, MMP-9, TIMP-1, and TIMP-2 was observed in the ECM of decidual tissue from RPL patients. Moreover, it was reported that the expression of MMP-2 was significantly lower and MMP-9 were significantly higher in patients with normal fetal chromosome than in abnormal.[55]

ECM in the ovary

Regulation of ECM in the ovary

In the ovary, collagen degradation is important in the process of ovulation. During luteal involution, activation of MMPs is crucial for protein depletion. MMP-2 may degrade collagen type IV, while membrane-type 1 MMP (MMP-14) may activate MMP-2 and degrade collagen type I, III, and IV.[56] MMP-14 and MMP-2 are also expressed in developing human ovarian follicles, and MMP-2 is present in the follicular fluid.[57] MMP-9 was also detected in the follicular fluid with a lower amount of activity than MMP-2, and a decrease of MMP-9 was observed during the change in follicle shape prior to ovulation.[58] A significant increase of MMP-10 expression in human granulosa cells between the pre and late-ovulatory phases was also reported.[59] Besides, HA is of great importance in ovarian ECM, which plays a role in regulating ovarian stiffness along with collagen. The content of HA in ovarian ECM is regulated by hyaluronidase (Hyal1) and hyaluronate synthase (Has3) and decreases with age.[60]

Change of ECM during ovulation

The composition of follicular basal lamina (mainly composed of collagen type IV and laminin) dynamically changes during ovulation. As the oocyte grows and the granulosa cells proliferate, the basal lamina expands, and the zona pellucida matrix develops around the oocyte.[61] The basal lamina is degraded on ovulation but preserved if atresia occurs.[62] Besides, an HA-abundant matrix forms within the oocyte complex during ovulation and is critical for fertility.[61]

Pathological states in the ovary

In patients with ovarian dysfunction, such as premature ovarian failure (POF) and polycystic ovarian syndrome (PCOS), their ovarian ECM may also change. POF refers to the loss of ovarian functions before the age of 40,[63] and is the outcome of ovarian fibrosis, which can be caused by surgery, inflammation, and abnormal immune reaction and undergoes ECM deposition.[64] PCOS results from an imbalance in female sex hormones, leading to cysts in the ovarian antral follicles.[65] Studies have shown that the circulating levels of MMP-2 and -9 increase in women with PCOS, which may be associated with abnormal basement membrane rupture or follicular atresia.[66]

ECM-DERIVED MATERIALS

Components of ECM

Inspired by the properties of ECM, including its composition, function, and metabolism, research on ECM-derived materials is flourishing. A summary of the main applications of ECM-derived materials in reproductive medicine is presented in Table 2. Among others, the components of ECM are the simplest ECM-derived materials, such as collagen and HA.

Table 2: Summary of applications of common ECM-derived materials
Materials Introduction Main applications
Components of ECM Basic ECM-derived materials
Collagen The most important protein of ECM Endometrial scaffold[67-69]
Coating for 2D follicle culture[70]
Fibronectin A protein of ECM Coating for 2D follicle culture[70]
Laminin A protein of ECM Coating for 2D follicle culture[70]
HA The most important GAG of ECM Adherence compound for ET[71]
Decellularized ECM A scaffold obtained by removing all the cells of tissues from their ECM Coating for MSCs culture[72]
Uterine scaffold[73-75]
Ovarian scaffold[76,77]
ECM-derived hydrogel An appropriate 3D culture media
Matrigel Composed of various ECM components 3D follicle culture[70]
Transplantable artificial ovary[78]
Collagen hydrogel Hydrogel mainly composed of collagen 3D follicle culture[79]
HA hydrogel Hydrogel mainly composed of HA 3D follicle culture[80]
ECM: extracellular matrix; HA: hyaluronic acid; GAG: glycosaminoglycan; ET: embryo transfer; 3D: 3-dimensional; 2D: 2-dimensional.

Collagen is a protein characterized by a distinctive triple helix super-secondary structure of three polypeptide chains.[9] As part of the ECM, collagen supports cell growth and contributes to mechanical resilience. Despite the advantages of being renewable, biocompatible, and biodegradable, collagen has limitations as a biomaterial that prevent its application, such as relatively weak mechanical strength and biological stability. Crosslinking is the joining of two or more molecules by a covalent bond, either intramolecularly or intermolecularly, and the reagents used for crosslinking are called crosslinkers.[81] Crosslinking is an approach to overcome these limitations for collagens and other biomaterials.[82,83] However, cross-linking can make polymeric materials less biodegradable and increase toxicity.[84] Therefore, it is suggested that more suitable crosslinking methods be developed.[85]

HA is an anionic, non-sulfated GAG composed of a stable oligosaccharide that consists of D-glucuronide and N-acetylglucosamine groups through glycosidic bonds.[10] HA’s properties, including cellular communication, healing and scar-forming, mediation of inflammation, connection, and hydration capacity, can be improved by crosslinking for tissue engineering.[10,86,87] HA is also a common crosslinker.[88] Besides, a high concentration of HA has been used in embryo transfer as an adherence compound, which may improve the outcomes of IVF/ICSI.[71]

Decellularized ECM

The decellularized ECM (dECM) is obtained by removing the cellular components of different tissues and organs from their ECM scaffold using mechanical, chemical, and enzymatic treatments.[89] The purpose of ECM decellularization is to preserve the components and structure of ECM while removing xenogeneic cells to avoid immune reactions.[90] The mechanical strength and stability of dECM scaffolds can be improved through crosslinking to produce more material products, such as hydrogels.[91] The dECM can be recellularized for in vivo and in vitro studies. Mesenchymal stem cells (MSCs) are a heterogeneous population of stromal stem cells with regeneration properties and immunomodulation properties.[92] However, their poor viability at the transplantation site limits their clinical application.[93] The dECM can provide a better environment for the MSCs, promotes their proliferation, and preserve stem cell properties.[94,95] This combination may overcome a series of challenges.[96]

ECM-derived Hydrogel

Hydrogels are a class of 3D water-swollen networks of polymers that are assembled by physical or chemical crosslinking processes. Among the components of ECM, collagen is commonly used as a natural material for hydrogel. Hyaluronic acid is also a common material for hydrogels, but most of the hyaluronic acid used in hydrogels is semi-synthetic.[97] Hydrogels have special physiochemical properties, including water-swelling, porous structures, and self-healing. Naturally, derived hydrogels usually have good biocompatibility and biodegradability but poor mechanical properties, including rigidity and stretchability. Therefore, crosslinking mediated by glycation and enzyme are typically used to increase these mechanical properties.[98]

Hydrogel is an appropriate 3D culture media to explore the migration and proliferation of stem cells instead of using ECM directly.[99] It also has been applied in treating diseases, such as intrauterine adhesions, as physical barriers or delivery systems, therefore playing a role in promoting tissue repair, reducing infection, and delivering therapeutic drugs.[100]

APPLICATION OF ECM-DERIVED MATERIALS IN REPRODUCTIVE TISSUE ENGINEERING

Uterine tissue engineering

Uterine tissue engineering offers hope for patients with uterine infertility. The development of scaffolds is always the first and vital step, and scaffolds for reproductive tissue engineering have been explored.[101,102] ECM-derived materials, including natural, synthetic, and decellularized ECM materials, have been used.[103]

Tissue engineering dominated by a single natural or synthetic material usually focuses on the endometrium. In a study,[104] collagen type I obtained from bovine was used to construct a scaffold with thermal dehydration crosslinking. This scaffold was then loaded with human umbilical cord-derived MSCs (UC-MSCs) and used in an induced murine uterine defect model. So far, clinical trials on humans also have been conducted.[68,69] A phase I clinical trial using allogeneic UC-MSCs on collagen scaffolds for patients with uterine adhesion reported benefits to patients’ endometrial condition, as well as successful pregnancy and delivery.[68]

The design and application of dECM scaffolds, which usually involve the full-thickness uterine tissue, is a different strategy. In one study,[73] rat uteri were decellularized using detergents (including sodium dodecyl sulfate, Triton X 100, and sterile phosphate buffered saline) to produce dECM scaffolds, which were then placed onto a partially excised uterus, realizing recellularization and regeneration in vivo and even successful pregnancy. Studies have shown that the sampling site of ECM, the methods to decellularize, and the transplanting orientation of the scaffold are all critical to the outcome.[74,75]

Ovarian tissue engineering

In recent years, due to health and social factors, the demand for fertility preservation has increased dramatically.[105] In such cases, the construction of an artificial ovary is an idea that demands a higher level of material science.

ECM coating has been used for follicle culture, but it has been found that the structure of follicles is better preserved in the 3D matrix.[70] One study[79] used a collagen hydrogel core (softer) and a degradable alginate hydrogel shell (harder) to encapsulate follicles of deer mice, mimicking the medulla and cortex of the ovary respectively, and successfully created a 3D culture environment. Another study[80] reported an HA-alginate hydrogel as a promising material for in vitro mouse follicle culture. For in vivo experimentation, an alginate-Matrigel matrix containing isolated ovarian cells has been designed to construct a transplantable artificial ovary. Seven days after being grafted into the mouse model, degradation of the matrix scaffold and angiogenesis around the follicles were observed.[78] However, finding a matrix to cultivate and transplant isolated human follicles is still challenging. The dECM seems to be a promising material for the scaffold of the transplantable artificial ovary. In a recent study,[76] donated cryopreserved ovarian tissues were decellularized and reseeded with human ovarian stromal cells and preantral follicles. These artificial ovaries were grafted into immunodeficient mice, with 5 out of 20 follicles managing to be recovered. It is worth mentioning that the donors of ovarian tissue include patients with malignant tumors, while no malignant cells were found in the grafted tissues. Another study[77] reported clinical cases that used dECM scaffold to help transplant frozen-banked ovarian tissue back into patients. Follicular development was observed and both of the patients were able to conceive through IVF-ET, with one having already successfully delivered.

CONCLUSIONS AND PERSPECTIVES

This review focuses on the regulation of ECM in the uterus and ovary, and discusses the progress of ECM-derived materials in uterine and ovarian tissue engineering. ECM is involved in and plays an important role in pregnancy and ovulation. However, most current studies on ECM and infertility seem to focus on mechanistic hypotheses, while no clear pathways have been investigated yet. However, the importance of ECM as a tissue scaffold and an environment for cellular activity is undeniable. Therefore, ECM-derived materials are critical in uterine and ovarian tissue engineering, providing new ideas for solving reproductive medicine clinical problems from materials science and regenerative medicine perspectives.

On the one hand, ECM-derived materials can be used for organ repair and reconstruction, which are novel therapeutic strategies for infertility. We follow the progress of such research and are inspired by several clinical trials. These translational medicine results bridge tissue engineering and clinical medicine, providing experience to solve clinical problems. On the other hand, the mechanical and biological stability of ECM-derived materials is not satisfactory enough, for ECM has a complex structure and can be regulated by multiple factors in vivo. This is also a challenge in the translation from in vitro to in vivo and from basic to clinical research. Therefore, modifying strategies to improve the properties of ECM-derived materials while preserving their advantages deserve further investigation—for example, the crosslinking and decellularization methods for dECM. Recently, 3D-printing technology has greatly broadened the horizon of tissue engineering and regenerative medicine. Developing bio-ink of ECM-derived materials with better performance for reproductive medicine is also a promising research direction.

In future research, a deeper understanding of the role played by ECM in infertility, leading to targeted modification of ECM-derived materials to enable better applications in reproductive medicine, would be a bright perspective to promote the combination and co-development of reproductive medicine and materials science.

DECLARATIONS

Author Contributions

Zhengyan Hu: Conceptualization, Methodology, Investigation, Writing—Original draft preparation. Rui Gao: Investigation. Hanxiao Chen: Writing—Reviewing and Editing. Minqi Chen: Investigation. Lang Qin: Conceptualization, Supervision.

Conflict of Interest

None declared.

REFERENCES

  1. Borght M, Wyns C. Fertility and infertility: definition and epidemiology. Clin Biochem 2018;62:2-10.    DOI: 10.1016/j.clinbiochem.2018.03.012    PMID: 29555319
  2. Steptoe PC, Edwards RG. Birth after the reimplantation of a human embryo. Lancet 1978;2(8085):366.    DOI: 10.1016/s0140-6736(78)92957-4    PMID: 79723
  3. Palermo G, Joris H, Devroey P, van Steirteghem AC. Pregnancies after intracytoplasmic injection of single spermatozoon into an oocyte. Lancet 1992;340(8810):17-18.    DOI: 10.1016/0140-6736(92)92425-f    PMID: 1351601
  4. Karamanos NK, Theocharis AD, Piperigkou Z, et al. A guide to the composition and functions of the extracellular matrix. FEBS J 2021;288(24):6850-6912.    DOI: 10.1111/febs.15776    PMID: 33605520
  5. O'Connor BB, Pope BD, Peters MM, Ris-Stalpers C, Parker KK. The role of extracellular matrix in normal and pathological pregnancy: future applications of microphysiological systems in reproductive medicine. Exp Biol Med 2020;245(13):1163-1174.    DOI: 10.1177/1535370220938741    PMID: 32640894
  6. Chen C. Pregnancy after human oocyte cryopreservation. Lancet 1986;1(8486):884-886.    DOI: 10.1016/s0140-6736(86)90989-x    PMID: 2870356
  7. Bedaiwy MA, Shahin AY, Falcone T. Reproductive organ transplantation: advances and controversies. Fertil Steril 2008;90(6):2031-2055.    DOI: 10.1016/j.fertnstert.2008.08.009    PMID: 18930221
  8. Badylak SF, Freytes DO, Gilbert TW. Extracellular matrix as a biological scaffold material: structure and function. Acta Biomater 2009;5(1):1-13.    DOI: 10.1016/j.actbio.2008.09.013    PMID: 18938117
  9. Gelse K, Pöschl E, Aigner T. Collagens—structure, function, and biosynthesis. Adv Drug Deliv Rev 2003;55(12):1531-1546.    DOI: 10.1016/j.addr.2003.08.002    PMID: 14623400
  10. Vasvani S, Kulkarni P, Rawtani D. Hyaluronic acid: a review on its biology, aspects of drug delivery, route of administrations and a special emphasis on its approved marketed products and recent clinical studies. Int J Biol Macromol 2020;151:1012-1029.    DOI: 10.1016/j.ijbiomac.2019.11.066    PMID: 31715233
  11. Wen Q, Mithieux SM, Weiss AS. Elastin biomaterials in dermal repair. Trends Biotechnol 2020;38(3):280-291.    DOI: 10.1016/j.tibtech.2019.08.005    PMID: 31870589
  12. Vindin H, Mithieux SM, Weiss AS. Elastin architecture. Matrix Biol 2019;84:4-16.    DOI: 10.1016/j.matbio.2019.07.005    PMID: 31301399
  13. Almine JF, Bax DV, Mithieux SM, et al. Elastin-based materials. Chem Soc Rev 2010;39(9):3371-3379.    DOI: 10.1039/b919452p    PMID: 20449520
  14. Aumailley M. The laminin family. Cell Adh Migr 2013;7(1):48-55.    DOI: 10.4161/cam.22826    PMID: 23263632
  15. Yurchenco PD, McKee KK, Reinhard JR, Rüegg MA. Laminin-deficient muscular dystrophy: molecular pathogenesis and structural repair strategies. Matrix Biol 2018;71–72:174-187.    DOI: 10.1016/j.matbio.2017.11.009    PMID: 29191403
  16. Patten J, Wang K. Fibronectin in development and wound healing. Adv Drug Deliv Rev 2021;170:353-368.    DOI: 10.1016/j.addr.2020.09.005    PMID: 32961203
  17. Werb Z. ECM and cell surface proteolysis: regulating cellular ecology. Cell 1997;91(4):439-442.    DOI: 10.1016/s0092-8674(00)80429-8    PMID: 9390552
  18. Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ Res 2003;92(8):827-839.    DOI: 10.1161/01.RES.0000070112.80711.3D    PMID: 12730128
  19. Edwards DR, Handsley MM, Pennington CJ. The ADAM metalloproteinases. Mol Aspects Med 2008;29(5):258-289.    DOI: 10.1016/j.mam.2008.08.001    PMID: 18762209
  20. Mead TJ, Apte SS. ADAMTS proteins in human disorders. Matrix Biol 2018;71–72:225-239.    DOI: 10.1016/j.matbio.2018.06.002    PMID: 29885460
  21. Gross J, Lapiere CM. Collagenolytic activity in amphibian tissues: a tissue culture assay. Proc Natl Acad Sci U S A 1962;48(6):1014-1022.    DOI: 10.1073/pnas.48.6.1014    PMID: 13902219
  22. Cui N, Hu M, Khalil RA. Biochemical and biological attributes of matrix metalloproteinases. Prog Mol Biol Transl Sci 2017;147:1-73.    DOI: 10.1016/bs.pmbts.2017.02.005    PMID: 28413025
  23. Zitka O, Kukacka J, Krizkova S, et al. Matrix metalloproteinases. Curr Med Chem 2010;17(31):3751-3768.    DOI: 10.2174/092986710793213724    PMID: 20846107
  24. Arpino V, Brock M, Gill SE. The role of TIMPs in regulation of extracellular matrix proteolysis. Matrix Biol 2015;44–46:247-254.    DOI: 10.1016/j.matbio.2015.03.005    PMID: 25805621
  25. Ågren MS, Schnabel R, Christensen LH, Mirastschijski U. Tumor necrosis factor-α-accelerated degradation of type I collagen in human skin is associated with elevated matrix metalloproteinase (MMP)-1 and MMP-3 ex vivo. Eur J Cell Biol 2015;94(1):12-21.    DOI: 10.1016/j.ejcb.2014.10.001    PMID: 25457675
  26. Mirastschijski U, Lupše B, Maedler K, et al. Matrix metalloproteinase-3 is key effector of TNF-α-induced collagen degradation in skin. Int J Mol Sci 2019;20(20):5234.    DOI: 10.3390/ijms20205234    PMID: 31652545
  27. Li X, Guo L, Yang X, et al. TGF-β1-induced connexin43 promotes scar formation via the Erk/MMP-1/collagen III pathway. J Oral Rehabil 2020;47(Suppl 1):99-106.    DOI: 10.1111/joor.12829    PMID: 31175668
  28. Chen LC, Shibu MA, Liu CJ, et al. ERK1/2 mediates the lipopolysaccharide-induced upregulation of FGF-2, uPA, MMP-2, MMP-9 and cellular migration in cardiac fibroblasts. Chem Biol Interact 2019;306:62-69.    DOI: 10.1016/j.cbi.2019.04.010    PMID: 30980805
  29. Salamonsen LA, Zhang J, Hampton A, Lathbury L. Regulation of matrix metalloproteinases in human endometrium. Hum Reprod 2000;15(Suppl 3):112-119.    DOI: 10.1093/humrep/15.suppl_3.112    PMID: 11041227
  30. Salamonsen LA, Woolley DE. Matrix metalloproteinases in normal menstruation. Hum Reprod 1996;11(Suppl 2):124-133.    DOI: 10.1093/humrep/11.suppl_2.124    PMID: 8982754
  31. Obokata A, Watanabe J, Nishimura Y, Arai T, Kawaguchi M, Kuramoto H. Significance of matrix metalloproteinase-7[correction of matrix metalloproteinase-2],-11 and tissue inhibitor of metalloproteinase-1 expression in normal, hyperplastic and neoplastic endometrium. Anticancer Res 2007;27(1A):95-105.    PMID: 17352221
  32. Chakraborti S, Mandal M, Das S, Mandal A, Chakraborti T. Regulation of matrix metalloproteinases: an overview. Mol Cell Biochem 2003;253(1):269-285.    DOI: 10.1023/a:1026028303196    PMID: 14619979
  33. Yoshii N, Hamatani T, Inagaki N, et al. Successful implantation after reducing matrix metalloproteinase activity in the uterine cavity. Reprod Biol Endocrinol 2013;11:37.    DOI: 10.1186/1477-7827-11-37    PMID: 23663265
  34. Basu J, Agamasu E, Bendek B, et al. Correlation Between Placental Matrix Metalloproteinase 9 and Tumor Necrosis Factor-α Protein Expression Throughout Gestation in Normal Human Pregnancy. Reprod Sci 2018;25(4):621-627.    DOI: 10.1177/1933719117725819    PMID: 28820024
  35. Xu P, Alfaidy N, Challis JR. Expression of matrix metalloproteinase (MMP)-2 and MMP-9 in human placenta and fetal membranes in relation to preterm and term labor. J Clin Endocrinol Metab 2002;87(3):1353-1361.    DOI: 10.1210/jcem.87.3.8320    PMID: 11889208
  36. Wang W, Vilella F, Alama P, et al. Single-cell transcriptomic atlas of the human endometrium during the menstrual cycle. Nat Med 2020;26(10):1644-1653.    DOI: 10.1038/s41591-020-1040-z    PMID: 32929266
  37. Sternberg AK, Buck VU, Classen-Linke I, Leube RE. How mechanical forces change the human endometrium during the menstrual cycle in preparation for embryo implantation. Cells 2021;10(8):2008.    DOI: 10.3390/cells10082008    PMID: 34440776
  38. Schatz F, Guzeloglu-Kayisli O, Arlier S, Kayisli UA, Lockwood CJ. The role of decidual cells in uterine hemostasis, menstruation, inflammation, adverse pregnancy outcomes and abnormal uterine bleeding. Hum Reprod Update 2016;22(4):497-515.    DOI: 10.1093/humupd/dmw004    PMID: 26912000
  39. Burrows TD, King A, Smith SK, Loke YW. Human trophoblast adhesion to matrix proteins: inhibition and signal transduction. Hum Reprod 1995;10(9):2489-2500.    DOI: 10.1093/oxfordjournals.humrep.a136329    PMID: 8530696
  40. Talaulikar V, Kronenberger K, Bax BE, Moss R, Manyonda I. Differences in collagen ultrastructure of human first trimester decidua basalis and parietalis: implications for trophoblastic invasion of the placental bed. J Obstet Gynaecol Res 2014;40(1):80-88.    DOI: 10.1111/jog.12127    PMID: 23937248
  41. Tanaka T, Wang C, Umesaki N. Remodeling of the human endometrial epithelium is regulated by laminin and type IV collagen. Int J Mol Med 2009;23(2):173-180.    PMID: 19148540
  42. Yang ZS, Pan HY, Shi WW, et al. Regulation and function of laminin A5 during mouse and human decidualization. Int J Mol Sci 2021;23(1):199.    DOI: 10.3390/ijms23010199    PMID: 35008625
  43. Sanders B. Uterine factors and infertility. J Reprod Med 2006;51(3):169-176.    PMID: 16674011
  44. Liu KE, Hartman M, Hartman A. Management of thin endometrium in assisted reproduction: a clinical practice guideline from the Canadian Fertility and Andrology Society. Reprod Biomed Online 2019;39(1):49-62.    DOI: 10.1016/j.rbmo.2019.02.013    PMID: 31029557
  45. Berman JM. Intrauterine adhesions. Semin Reprod Med 2008;26(4):349-355.    DOI: 10.1055/s-0028-1082393    PMID: 18756412
  46. Nejat EJ, Zapantis G, Rybak EA, Meier UT. It’s time to pay attention to the endometrium, including the nucleolar channel system. Fertil Steril 2011;96(6):e165;authorreplye166.    DOI: 10.1016/j.fertnstert.2011.09.053    PMID: 22000912
  47. Jiang P, Tang X, Wang H, et al. Collagen-binding basic fibroblast growth factor improves functional remodeling of scarred endometrium in uterine infertile women: a pilot study. Sci China Life Sci 2019;62(12):1617-1629.    DOI: 10.1007/s11427-018-9520-2    PMID: 31515729
  48. Cimadomo D, Craciunas L, Vermeulen N, Vomstein K, Toth B. Definition, diagnostic and therapeutic options in recurrent implantation failure: an international survey of clinicians and embryologists. Hum Reprod 2021;36(2):305-317.    DOI: 10.1093/humrep/deaa317    PMID: 33313697
  49. Turpeenniemi-Hujanen T, Feinberg RF, Kauppila A, Puistola U. Extracellular matrix interactions in early human embryos: implications for normal implantation events. Fertil Steril 1995;64(1):132-138.    PMID: 7789549
  50. Fouladi-Nashta AA, Raheem KA, Marei WF, Ghafari F, Hartshorne GM. Regulation and roles of the hyaluronan system in mammalian reproduction. Reproduction 2017;153(2):R43-R58.    DOI: 10.1530/REP-16-0240    PMID: 27799626
  51. Zhang C, Duan E, Cao Y, Jiang G, Zeng G. Effect of 32/67 kDa laminin-binding protein antibody on mouse embryo implantation. J Reprod Fertil 2000;119(1):137-142.    DOI: 10.1530/jrf.0.1190137    PMID: 10864823
  52. Klemmt PA, Carver JG, Koninckx P, McVeigh EJ, Mardon HJ. Endometrial cells from women with endometriosis have increased adhesion and proliferative capacity in response to extracellular matrix components: towards a mechanistic model for endometriosis progression. Hum Reprod 2007;22(12):3139-3147.    DOI: 10.1093/humrep/dem262    PMID: 17921481
  53. Dimitriadis E, Menkhorst E, Saito S, Kutteh WH, Brosens JJ. Recurrent pregnancy loss. Nat Rev Dis Primers 2020;6(1):98.    DOI: 10.1038/s41572-020-00228-z    PMID: 33303732
  54. Jerzak M, Rechberger T, Górski A. Intravenous immunoglobulin therapy influences T cell adhesion to extracellular matrix in women with a history of recurrent spontaneous abortions. Am J Reprod Immunol 2000;44(6):336-341.    DOI: 10.1111/j.8755-8920.2000.440603.x    PMID: 11200811
  55. Ozawa F, Mori R, Obayashi Y, et al. Clinical significance of MMPs (matrix metalloproteinases) and TIMPs (tissue inhibitors of metalloproteinases) with recurrent miscarriage. Nagoya: The 71st Annual Congress of the Japan Society of Obstetrics and Gynecology, 2019.
  56. Endo T, Kiya T, Goto T, et al. Significance of matrix metalloproteinases in the pathophysiology of the ovary and uterus. Reprod Med Biol 2006;5(4):235-243.    DOI: 10.1111/j.1447-0578.2006.00147.x    PMID: 29699252    PMCID: PMC5904581
  57. Vos MC, van der Wurff AA, Last JT, et al. Immunohistochemical expression of MMP-14 and MMP-2, and MMP-2 activity during human ovarian follicular development. Reprod Biol Endocrinol 2014;12:12.    DOI: 10.1186/1477-7827-12-12    PMID: 24485069
  58. Riley SC, Thomassen R, Bae SE, Leask R, Pedersen HG, Watson ED. Matrix metalloproteinase-2 and-9 secretion by the equine ovary during follicular growth and prior to ovulation. Anim Reprod Sci 2004;81(3–4):329-339.    DOI: 10.1016/j.anireprosci.2003.10.011    PMID: 14998657
  59. McCord L, Park E, Jo M, Brannstrom M, Curry TE. Expression of matrix metalloproteinase 10 (Mmp10) during the periovulatory period. Biol Reprod 2010;83(Suppl_1):642.
  60. Amargant F, Manuel SL, Tu Q, et al. Ovarian stiffness increases with age in the mammalian ovary and depends on collagen and hyaluronan matrices. Aging Cell 2020;19(11):e13259.    DOI: 10.1111/acel.13259    PMID: 33079460
  61. Irving-Rodgers HF, Rodgers RJ. Extracellular matrix in ovarian follicular development and disease. Cell Tissue Res 2005;322(1):89-98.    DOI: 10.1007/s00441-005-0042-y    PMID: 16158327
  62. Irving-Rodgers HF, Mussard ML, Kinder JE, Rodgers RJ. Composition and morphology of the follicular basal lamina during atresia of bovine antral follicles. Reproduction 2002;123(1):97-106.    PMID: 11869191
  63. Jankowska K. Premature ovarian failure. Menopause Rev 2017;16(2):51-56.    DOI: 10.5114/pm.2017.68592    PMID: 28721130
  64. Zhou F, Shi LB, Zhang SY. Ovarian fibrosis: a phenomenon of concern. Chin Med J 2017;130(3):365-371.    DOI: 10.4103/0366-6999.198931    PMID: 28139522
  65. Patel S. Polycystic ovary syndrome (PCOS), an inflammatory, systemic, lifestyle endocrinopathy. J Steroid Biochem Mol Biol 2018;182:27-36.    DOI: 10.1016/j.jsbmb.2018.04.008    PMID: 29678491
  66. Lewandowski KC, Komorowski J, O'Callaghan CJ, et al. Increased circulating levels of matrix metalloproteinase-2 and-9 in women with the polycystic ovary syndrome. J Clin Endocrinol Metab 2006;91(3):1173-1177.    DOI: 10.1210/jc.2005-0648    PMID: 16338908
  67. Xu L, Ding L, Wang L, et al. Umbilical cord-derived mesenchymal stem cells on scaffolds facilitate collagen degradation via upregulation of MMP-9 in rat uterine scars. Stem Cell Res Ther 2017;8(1):84.    DOI: 10.1186/s13287-017-0535-0    PMID: 28420433
  68. Cao Y, Sun H, Zhu H, et al. Allogeneic cell therapy using umbilical cord MSCs on collagen scaffolds for patients with recurrent uterine adhesion: a phase I clinical trial. Stem Cell Res Ther 2018;9(1):192.    DOI: 10.1186/s13287-018-0904-3    PMID: 29996892
  69. Zhang Y, Shi L, Lin X, et al. Unresponsive thin endometrium caused by Asherman syndrome treated with umbilical cord mesenchymal stem cells on collagen scaffolds: a pilot study. Stem Cell Res Ther 2021;12(1):420.    DOI: 10.1186/s13287-021-02499-z    PMID: 34294152
  70. Oktem O, Oktay K. The role of extracellular matrix and activin-A in in vitro growth and survival of murine preantral follicles. Reprod Sci 2007;14(4):358-366.    DOI: 10.1177/1933719107303397    PMID: 17644808
  71. Heymann D, Vidal L, Or Y, Shoham Z. Hyaluronic acid in embryo transfer media for assisted reproductive technologies. Cochrane Database Syst Rev 2020;9(9):CD007421.    DOI: 10.1002/14651858.CD007421.pub4    PMID: 32876946
  72. Shakouri-Motlagh A, O'Connor AJ, Brennecke SP, Kalionis B, Heath DE. Native and solubilized decellularized extracellular matrix: a critical assessment of their potential for improving the expansion of mesenchymal stem cells. Acta Biomater 2017;55:1-12.    DOI: 10.1016/j.actbio.2017.04.014    PMID: 28412553
  73. Miyazaki K, Maruyama T. Partial regeneration and reconstruction of the rat uterus through recellularization of a decellularized uterine matrix. Biomaterials 2014;35(31):8791-8800.    DOI: 10.1016/j.biomaterials.2014.06.052    PMID: 25043501
  74. Miki F, Maruyama T, Miyazaki K, et al. The orientation of a decellularized uterine scaffold determines the tissue topology and architecture of the regenerated uterus in rats. Biol Reprod 2019;100(5):1215-1227.    DOI: 10.1093/biolre/ioz004    PMID: 30649202
  75. Padma AM, Tiemann TT, Alshaikh AB, Akouri R, Song MJ, Hellström M. Protocols for rat uterus isolation and decellularization: applications for uterus tissue engineering and 3D cell culturing. Methods Mol Biol 2018;1577:161-175.    DOI: 10.1007/7651_2017_60    PMID: 28776178
  76. Pors SE, Ramløse M, Nikiforov D, et al. Initial steps in reconstruction of the human ovary: survival of pre-antral stage follicles in a decellularized human ovarian scaffold. Hum Reprod 2019;34(8):1523-1535.    DOI: 10.1093/humrep/dez077    PMID: 31286144
  77. Oktay K, Bedoschi G, Pacheco F, Turan V, Emirdar V. First pregnancies, live birth, and in vitro fertilization outcomes after transplantation of frozen-banked ovarian tissue with a human extracellular matrix scaffold using robot-assisted minimally invasive surgery. Am J Obstet Gynecol 2016;214(1):94.e1-94.e9.    DOI: 10.1016/j.ajog.2015.10.001    PMID: 26601616
  78. Vanacker J, Luyckx V, Dolmans MM, et al. Transplantation of an alginate-matrigel matrix containing isolated ovarian cells: first step in developing a biodegradable scaffold to transplant isolated preantral follicles and ovarian cells. Biomaterials 2012;33(26):6079-6085.    DOI: 10.1016/j.biomaterials.2012.05.015    PMID: 22658800
  79. Choi JK, Agarwal P, Huang H, Zhao S, He X. The crucial role of mechanical heterogeneity in regulating follicle development and ovulation with engineered ovarian microtissue. Biomaterials 2014;35(19):5122-5128.    DOI: 10.1016/j.biomaterials.2014.03.028    PMID: 24702961
  80. Jamalzaei P, Rezazadeh Valojerdi M, Montazeri L, Baharvand H. Applicability of hyaluronic acid-alginate hydrogel and ovarian cells for in vitro development of mouse preantral follicles. Cell J 2020;22(Suppl 1):49-60.    DOI: 10.22074/cellj.2020.6925    PMID: 32779433
  81. Arora B, Tandon R, Attri P, Bhatia R. Chemical crosslinking: role in protein and peptide science. Curr Protein Pept Sci 2017;18(9):946-955.    DOI: 10.2174/1389203717666160724202806    PMID: 27455969
  82. Koide T, Daito M. Effects of various collagen crosslinking techniques on mechanical properties of collagen film. Dent Mater J 1997;16(1):1-9.    DOI: 10.4012/dmj.16.1    PMID: 9549996
  83. Reddy N, Reddy R, Jiang Q. Crosslinking biopolymers for biomedical applications. Trends Biotechnol 2015;33(6):362-369.    DOI: 10.1016/j.tibtech.2015.03.008    PMID: 25887334
  84. Jiang Q, Reddy N, Yang Y. Cytocompatible cross-linking of electrospun zein fibers for the development of water-stable tissue engineering scaffolds. Acta Biomater 2010;6(10):4042-4051.    DOI: 10.1016/j.actbio.2010.04.024    PMID: 20438870
  85. Delgado LM, Bayon Y, Pandit A, Zeugolis DI. To cross-link or not to cross-link? Tissue Eng Part B Rev 2015;21(3):298-313.    DOI: 10.1089/ten.TEB.2014.0290    PMID: 25517923
  86. Kobayashi T, Chanmee T, Itano N. Hyaluronan: metabolism and function. Biomolecules 2020;10(11):1525.    DOI: 10.3390/biom10111525    PMID: 33171800
  87. Segura T, Anderson BC, Chung PH, Webber RE, Shull KR, Shea LD. Crosslinked hyaluronic acid hydrogels: a strategy to functionalize and pattern. Biomaterials 2005;26(4):359-371.    DOI: 10.1016/j.biomaterials.2004.02.067    PMID: 15275810
  88. Lou J, Stowers R, Nam S, Xia Y, Chaudhuri O. Stress relaxing hyaluronic acid-collagen hydrogels promote cell spreading, fiber remodeling, and focal adhesion formation in 3D cell culture. Biomaterials 2018;154:213-222.    DOI: 10.1016/j.biomaterials.2017.11.004    PMID: 29132046
  89. Ge F, Lu Y, Li Q, Zhang X. Decellularized extracellular matrices for tissue engineering and regeneration. Adv Exp Med Biol 2020;1250:15-31.    DOI: 10.1007/978-981-15-3262-7_2    PMID: 32601935
  90. Liu C, Pei M, Li Q, Zhang Y. Decellularized extracellular matrix mediates tissue construction and regeneration. Front Med 2022;16(1):56-82.    DOI: 10.1007/s11684-021-0900-3    PMID: 34962624
  91. Ma B, Wang X, Wu C, Chang J. Crosslinking strategies for preparation of extracellular matrix-derived cardiovascular scaffolds. Regen Biomater 2014;1(1):81-89.    DOI: 10.1093/rb/rbu009    PMID: 26816627    PMCID: PMC4669006
  92. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol 2008;8(9):726-736.    DOI: 10.1038/nri2395    PMID: 19172693
  93. Talovic M, Patel K, Schwartz M, Madsen J, Garg K. Decellularized extracellular matrix gelloids support mesenchymal stem cell growth and function in vitro. J Tissue Eng Regen Med 2019;13(10):1830-1842.    DOI: 10.1002/term.2933    PMID: 31306568
  94. Lai Y, Sun Y, Skinner CM, et al. Reconstitution of marrow-derived extracellular matrix. ex vivo 2010;19(7):1095-1107.    DOI: 10.1089/scd.2009.0217    PMID: 19737070
  95. Lin H, Yang G, Tan J, Tuan RS. Influence of decellularized matrix derived from human mesenchymal stem cells on their proliferation, migration and multi-lineage differentiation potential. Biomaterials 2012;33(18):4480-4489.    DOI: 10.1016/j.biomaterials.2012.03.012    PMID: 22459197
  96. Dunn A, Talovic M, Patel K, Patel A, Marcinczyk M, Garg K. Biomaterial and stem cell-based strategies for skeletal muscle regeneration. J Orthop Res 2019;37(6):1246-1262.    DOI: 10.1002/jor.24212    PMID: 30604468
  97. Caliari SR, Burdick JA. A practical guide to hydrogels for cell culture. Nat Methods 2016;13(5):405-414.    DOI: 10.1038/nmeth.3839    PMID: 27123816
  98. Cao H, Duan L, Zhang Y, Cao J, Zhang K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther 2021;6(1):426.    DOI: 10.1038/s41392-021-00830-x    PMID: 34916490
  99. Huang Q, Zou Y, Arno MC, et al. Hydrogel scaffolds for differentiation of adipose-derived stem cells. Chem Soc Rev 2017;46(20):6255-6275.    DOI: 10.1039/c6cs00052e    PMID: 28816316
  100. Lv H, Wu B, Song J, Wu W, Cai W, Xu J. Hydrogel, a novel therapeutic and delivery strategy, in the treatment of intrauterine adhesions. J Mater Chem B 2021;9(33):6536-6552.    DOI: 10.1039/d1tb01005k    PMID: 34324619
  101. Chan M, Keane T, Jones B, Smith JR, Stevens M, Saso S. Bioengineering approaches for female fertility preservation and infertility treatment. BJOG 2019;126(S2):196.
  102. Nikukar H. Regenerative medicine for female reproductive system. Accessed January 12, 2023. http://ijrm.ir/browse.php?mag_id=163&slc_lang=en&sid=1.
  103. Yoshimasa Y, Maruyama T. Bioengineering of the uterus. Reprod Sci 2021;28(6):1596-1611.    DOI: 10.1007/s43032-021-00503-8    PMID: 33826100
  104. Xin L, Lin X, Pan Y, et al. A collagen scaffold loaded with human umbilical cord-derived mesenchymal stem cells facilitates endometrial regeneration and restores fertility. Acta Biomater 2019;92:160-171.    DOI: 10.1016/j.actbio.2019.05.012    PMID: 31075515
  105. Donnez J, Dolmans MM. Fertility Preservation in Women. N Engl J Med 2017;377(17):1657-1665.    DOI: 10.1056/NEJMra1614676    PMID: 29069558

 

 

 

 

 

 

Similar Articles

You may also start an advanced similarity search for this article.