Next Article in Journal
Biotic and Abiotic Stressors in Plant Metabolism
Next Article in Special Issue
The Molecular Basis of Male Infertility in Obesity: A Literature Review
Previous Article in Journal
The Role of miRNAs in Childhood Acute Lymphoblastic Leukemia Relapse and the Associated Molecular Mechanisms
Previous Article in Special Issue
In Vitro Embryogenesis and Gastrulation Using Stem Cells in Mice and Humans
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Basis of Müllerian Agenesis Causing Congenital Uterine Factor Infertility—A Systematic Review

by
Rajani Dube
1,*,
Subhranshu Sekhar Kar
2,
Malay Jhancy
2 and
Biji Thomas George
3
1
Department of Obstetrics and Gynaecology, RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras al Khaimah P.O. Box 11172, United Arab Emirates
2
Department of Paediatrics and Neonatology, RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras al Khaimah P.O. Box 11172, United Arab Emirates
3
Department of General Surgery, RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras al Khaimah P.O. Box 11172, United Arab Emirates
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(1), 120; https://doi.org/10.3390/ijms25010120 (registering DOI)
Submission received: 17 October 2023 / Revised: 15 December 2023 / Accepted: 18 December 2023 / Published: 21 December 2023
(This article belongs to the Special Issue Molecular Studies in Endocrinology and Reproductive Biology)

Abstract

:
Infertility affects around 1 in 5 couples in the world. Congenital absence of the uterus results in absolute infertility in females. Müllerian agenesis is the nondevelopment of the uterus. Mayer–Rokitansky–Küster–Hauser (MRKH) syndrome is a condition of uterovaginal agenesis in the presence of normal ovaries and the 46 XX Karyotype. With advancements in reproductive techniques, women with MA having biological offspring is possible. The exact etiology of MA is unknown, although several genes and mechanisms affect the development of Müllerian ducts. Through this systematic review of the available literature, we searched for the genetic basis of MA. The aims included identification of the genes, chromosomal locations, changes responsible for MA, and fertility options, in order to offer proper management and counseling to these women with MA. A total of 85 studies were identified through searches. Most of the studies identified multiple genes at various locations, although the commonest involved chromosomes 1, 17, and 22. There is also conflicting evidence of the involvement of various candidate genes in the studies. The etiology of MA seems to be multifactorial and complex, involving multiple genes and mechanisms including various mutations and mosaicism.

1. Introduction

Infertility is defined by the World Health Organization (WHO) as a disease of the male or female reproductive system resulting in failure to achieve a pregnancy after 12 months or more of regular unprotected sexual intercourse [1]. It can be “Primary”, denoting those who have never become pregnant, or “Secondary”, depicting those with the inability to conceive after at least one previous pregnancy [2]. Infertility affects millions of people worldwide [3]. The prevalence of infertility can vary throughout the world, but generally affects around one in five couples [4]. Infertility can be caused by different factors in males, females, and can be combined or even unexplained [3]. Common causes in females are diseases of the ovaries, fallopian tubes, uterus, endocrinal, genital tract dysbiosis or combined, and differ from country to country [5]. Similarly, the cause can commonly be obstruction of the tract, testicular failure of spermatogenesis, poor sperm quality, or endocrinal in males [3]. Uterine factor infertility (UFI) is defined as an absent uterus (absolute UFI) or as a nonfunctional uterus (non-absolute UFI) [6]. Absolute UFI can be due to congenital absence of the uterus or due to a hysterectomy later [6]. UFI can affect about 1 in 500 women of reproductive age or up to 5% of females, although the exact data are unknown [7,8].
Embryologically, the uterus, fallopian tubes, and upper part of the vagina develop from Müllerian ducts (MD) (or the paramesonephric duct). The unfused cranial end of MDs remains separated and forms the fallopian tubes. The further cranial end of MDs from both sides fuse vertically to form the uterine body and cervix, and the caudal part of MDs fuse to form the upper portion of the vagina. The vagina then canalizes and fuses with the embryonic cloaca to complete the vaginal canal [9,10]. Ovarian development in utero, on the other hand, is a complex process. Four components of the ovaries, namely the surface epithelium, ovarian stroma, primordial germ cells, and sex cords, develop from the coelomic epithelium, sub-coelomic mesoderm, yolk sac endoderm, and invagination of cortical coelomic epithelium, respectively [11]. Favorable conditions for optimal development of the female genital system are the presence of both functional X chromosomes (46 XX) and the absence of the SRY gene [9]. Various agents can influence embryogenesis to cause structural abnormalities. While drugs like diethylstilbestrol (DES), ionizing radiations, and certain infections are known to be teratogenic, others may or may not have a role [12,13,14].
Müllerian agenesis (MA), (Müllerian aplasia, complete uterine aplasia) or Mayer–Rokitansky–Küster–Hauser (MRKH) syndrome, is a rare disorder with an incidence of 1 per 4500–5000 females [15]. Although not identical, MRKH and MA are interchangeably used in the literature. MRKH is further divided into two types. When there is an isolated MA, with normal ovaries and without involvement of other organ systems, it is called type 1 MRKH. MRKH type 2 includes an absent uterus along with abnormalities in the tubes, ovaries, and urinary system. Type 2 also includes a severe form called MURCS (Müllerian duct aplasia, unilateral renal agenesis, and cervicothoracic somite anomalies) [15,16]. Some consider MURCS a separate class and classify MRKH into typical (type 1), atypical (type 2), and MURCS [17]. The exact cause of MA is largely unknown due to the heterogeneity in the published literature. The disorder was long considered to be sporadic [15]. As interest in MRKH grew, there were many reported cases of familial occurrence. Hence, there emerged a subset of patients wherein an autosomal dominant inheritance with incomplete penetrance and variable expressivity was a definite probability [18,19]. Intrinsically, it is not possible to study maternal inheritance of cases with MA because of the nature of MA, which produces absolute infertility. However, it is possible now due to advances in fertility treatment which mean that having a biological child is highly possible [20]. Thus, there occurred a series of studies to search for candidate genes and specific genetic bases of MA. While earlier studies used microarrays or small gene panels/Sanger sequencing [21,22], the scenario has changed with the use of massively parallel sequencing, including whole-exome sequencing. Newer methods have opened wider opportunities for the search for genetic causes of MA [23]. Furthermore, the studies in discordant monozygotic twins with only one twin having MA support the role of environmental factors affecting the expressivity of the genetic abnormalities [24,25,26,27,28,29,30,31,32]. Hence, through this research, we aim to explore the genetic and molecular basis of MA. This will also help in counseling couples seeking newer treatment options to achieve parenthood of biologically related offspring.

2. Methods

2.1. Search Strategy

A systematic search of the electronic databases Pubmed, Scopus, Web of Science, Embase, and Google Scholar was carried out. Medical subject handling terms (MeSH) and free-text term keywords like Mayer–Rokitansky–Küster–Hauser syndrome, Müllerian agenesis, uterine aplasia, and uterine agenesis were used in combination with gene, genome, genetic or molecular to search for data in January 2023. Thereafter, manual updates were made on a weekly basis until 10 August 2023. There was no starting date for the search. The references of relevant studies were also hand-searched if they did not belong to these databases.

2.2. Eligibility Criteria

2.2.1. Inclusion Criteria

Studied were included if they fulfilled all the following criteria:
  • English language articles or where English translation is available;
  • Full-text articles reporting on human genes, genome, genetics, or molecular bases;
  • Containing information on Müllerian duct abnormality or Müllerian agenesis or Müllerian aplasia or uterine aplasia or Mayer–Rokitansky–Kuster–Hauser syndrome.
All eligible studies published before August 2023 were included for review.

2.2.2. Exclusion Criteria

Exclusions consisted of animal studies, duplicated studies, review articles, non-genetic studies, articles in languages other than English where translation was not possible, and studies where full-text articles were only available upon payment. Conference abstracts, expert opinions, and critical appraisals were also excluded.

2.3. Study Selection

Genetic analysis can be performed with various methods. Older studies relied on conventional comparative genomic hybridization (CGH), polymerase chain reaction (PCR), or fluorescent in situ hybridization (FISH), while current analyses are carried out using newer methods [33]. Data on the methods used in each study are also extracted, through the search. As each of these methods has distinct advantages, they are mentioned in brief.
Array–CGH (aCGH): This assay provides higher resolution than traditional CGH, and is used as an alternative means of genome-wide screening for copy number variations (CNVs). It combines traditional CGH principles with a microarray, and thus is not dependent on actively dividing cells. An aCGH can simultaneously detect aneuploidies, deletions, duplications, and amplifications of any locus represented on an array, as well as submicroscopic chromosomal abnormalities [34].
Whole-exon sequencing (WES): This allows variations in the protein-coding region of any gene to be identified, rather than in only a select few genes. It is an efficient method for detecting CNVs in potential candidate genes to identify the abnormalities possibly causing disease, as most known mutations that cause disease occur in exons. However, mutations in regulatory factors coded outside exons can be missed [35].
Whole-genome sequencing (WGS): This is a more advanced technique based on massive-genome sequencing, and is not dependent on the availability of predefined databases for comparison and matching [36]. It can detect abnormalities in a wide spectrum of genes. However, it is expensive and requires complex analysis [37].
After a thorough search of the databases, a total of 1308 results were retrieved. All the abstracts and study titles were screened, and duplicates were removed. Furthermore, there were a total of 1226 studies excluded, as they either did not fit the inclusion criteria, were only animal studies, included only vaginal agenesis, only abnormalities other than uterine agenesis, only gonadal dysgenesis, or did not explore the genetic basis of the disease. In a manual search of references, three case reports were found and included. Finally, 85 articles were included in the analysis. The Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) are in Figure 1.

2.4. Data Collection

All the authors (RD, SSK, MJ, BTG) reviewed all titles independently. The potential relevance of the studies to be included for review was agreed upon by discussion on a regular basis. Selected titles and abstracts were further screened between studies to reject the overlap of cases. Full-text copies of the selected papers were obtained and the relevant data were extracted. In the case of individual case reports, if the same patient was included in more than one study with similar characteristics and findings, only the report with a larger number of patients was included. As far as possible, single case reports were cross-checked with other reports from the same location and hospital. The decision on exclusion or inclusion was decided by discussion if the time frame and characteristics of the reported cases from the same center matched. The risk of bias was not assessed due to the nature of the studies.

3. Results and Discussion

The studies included only cohorts, case reports, case series, or retrospective analyses of laboratory samples. The genetic analysis was carried out using various methods. Older studies relied on conventional CGH, PCR, or FISH, while most of the newer analyses were performed using newer methods like aCGH, WES, or WGS [33]. To put the completeness of genetic analysis into context, the methods used in individual studies are considered in this review.

3.1. Genetic Basis of MA

A universally agreeable gene is yet to be found in the available evidence. There are elaborate investigations into candidate genes associated with MA. Out of the proposed genes, one or more were implicated in specific cohorts, but none were found in all. It is rational to propose that the genes or regulators of genes essentially involved in Müllerian duct development are most likely to be involved in MA [38]. The WNT signaling pathway genes (WNT4, WNT9B), the HOX family genes, LX1, HNF1B, and a few other candidate genes have been implicated by Mikhael et al. through WES, which was then confirmed by Sanger sequencing [39]. The copy number variants (CNVs) at different locations in chromosomes 1, 16, 17, and 22 were identified by this study [39] [Table 1]. A glossary of gene names is available as supplementary material.
To overcome the drawbacks of WES, WGS was used recently by Pan et al. In addition, to further strengthen the prior evidence on specific gene involvements, this study identified five de novo variants in nine patients with MA [41]. There are also certain case reports of the involvement of CFTR, β-catenin, and te HOXA10 gene in women with complete uterine aplasia. However, it was concluded that it is unlikely to be the causative factor [92,93,94]. In a recent study by Ragitha et al. using PCR sequencing of coding exons of the WNT4 gene in 32 women with MA and gonadal dysgenesis, single-nucleotide variations, nucleotide substitution in intronic regions not affecting the normal splicing mechanism, and synonymous polymorphism (c.861C > T; p.G287G, rs544988174) were reported. Hence, any indication of WNT4 involvement in MA was not found [76].
There are a few studies exploring the inheritance of MA in families. It was not conclusively found to have a particular inheritance pattern, although an autosomal dominant trait was suggested in a few and refuted in others [15,70,80,95,96]. The challenges were incomplete family tree availability or of a particular genetic basis. Analyzing the specific genetic composition of monozygotic twins is very helpful in the identification of genetic contributions to the pathophysiology of diseases. When a single embryo divides into two after fertilization, the resultant pregnancy is called a monozygotic twin pregnancy. As they have developed from a single embryo, it is assumed that they share identical genetic composition. Studying the genetic pattern of monozygotic twins where only one has a condition gives us an insight into additional causative factors responsible for occurrences like de novo dominant mutations, or somatic mutations in the specific tissue. In a recent study on five pairs of monozygotic twins discordant for MA, the uterine tissue remnants and blood were studied using WGS [53]. They reported a mosaic variant in ACTR3B. This variant was absent in the blood of the normal twin, had low and high allele frequency in the blood, and affected tissue of the twin with MA, respectively [53]. Few of the studies elaborated on transcriptome analysis of endometrial samples from the rudimentary tissues. In a previous study of 35 sporadic patients with MRKH, perturbations in endometrial transcriptomes were described [96] This study in uterine remnants using RNA sequencing demonstrated a large number of upregulated (1236 in MRKH type 1 and 801 in MRKH type 2) and downregulated (670 in MRKH type 1 and 373 in MRKH type 2) genes associated with MRKHS [97]. It was also found that genes encoding for estrogen receptor 1 were perturbed in a few other studies [25,83,84]. Analysis of endometrial tissue in monozygotic twins also showed similar perturbations in a recent study by Buchert et al. [53].
In a recent study by Brakta et al. 2023, genetic analysis using optical genome mapping in 87 women with MRKH and available parents revealed 14 structural variants in 17/87 (19.5%). These included deletions (n = 7), duplications (n = 3), one new translocation t(7;14)(q32;q32) (n = 5), a previously identified translocation-t(3;16)(p22.3;p13.3), and aneuploidies (n = 2). They also reported mosaicism in three cases for trisomy 12, a 7;14 translocation, and 45,X (75%)/46,XX (25%). It was concluded that the exact mechanism for MA may be mosaicisms [98]. In another study by Brendan et al. in eight individuals with MRKH, WES was used for analysis. The study reported multiple damaging and potentially damaging changes in more than one woman involving chromosomes 1, 3, 4, 7, 8, 11, 12, 20, and 22 [43]. Furthermore, few of the studies have reported no abnormality in the homeobox gene, the PAX2, WNT4, GALT, AMH, and AMHR genes, copy number changes, and AMH promotor sequence variations [17,99,100,101,102,103,104,105,106,107,108,109]. Another interesting study has reported a testis-specific protein 1-Y-linked (TSPY) gene in two women out of six with MRKHS and the 46XX karyotype [110]. Similarly, the level of galactose-1-phosphate uridyl transferase (GALT) was found to be lower in erythrocytes of women with vaginal agenesis, and two variants of the GALT gene were detected in another study [111]. This suggests that multiple genes and multiple mechanisms may be involved in the pathogenesis of MA.

3.2. Mechanisms of Genetic Changes

The development of female genital organs is a complex process and is influenced by the interplay of genetic, hormonal, and environmental factors. To understand the basis of genetic abnormalities resulting in MA, a brief overview of the development of MDs is essential.
During embryonic development, in both male and female fetuses, the gonadal ridges have the capacity to form either the testis or ovary until 6 weeks after conception. In the gonadal ridges, the supporting-cell lineage derived from the multipotent somatic progenitor cells is programmed to include pre-granulosa (WNT4, RSPO1, FST, and CTNNB1) genes in XX fetuses [112,113].
Müllerian (paramesonephric) ducts that give rise to most of the female reproductive tract arise around 5–6 weeks of gestation as a cleft lined by the coelomic epithelium in the urogenital ridge. Further development occurs in phases [114,115]. At first, there is a thickening of the coelomic epithelium along with expression of LHX1, and anti-Müllerian hormone receptor type II (AMHR2) [114,116,117,118]. DACH1 and DACH2 are transcriptional co-factors that act by regulating the expression of LHX1 and WNT7A and are required for the formation of MD [119,120]. In the next phase, these primordial Müllerian cells invaginate from the coelomic epithelium to reach the Wolffian duct. WNT4 expression in the mesonephric mesenchyme is essential for the Müllerian duct progenitor cells to begin invagination [116,121]. The last is the elongation phase, which begins when the invaginating tip of the Müllerian duct contacts the Wolffian duct. There is then proliferation and caudal migration of cells. There is continued elongation of MD, which eventually fuses centrally close to the urogenital sinus. MD then establishes apico-basal characteristics and develops into an epithelial tube that gives rise to the endometrium, and the surrounding mesenchyme differentiates into the myometrium of the uterus and Fallopian tubes [114,118]. The Wolffian duct plays an important role in the growth of MD, by supplying WNT9B secretion [122]. LIM1 or PAX2 are transcription factors contributing to MD growth.
SOX9 has an important role in the regression of the MDs. In male fetuses, the SRY (sex-determining region on the Y) gene encodes the transcription factor SOX9, which plays a vital role in gonadal differentiation. Upregulation of the expression of SOX9 in normal male development causes the development of Wolffian duct and degeneration of MDs upregulating the expression of anti-Müllerian hormone (AMH), and results in the downregulation of WNT4 expression [123,124,125,126,127,128]. Other members of the SOX family and various other factors act through upregulation or downregulation of SOX9 to control MD development. SOX3 can induce SOX9 expression, and SOX8 and SOX10 upregulate SOX9 expression [129]. Similarly, Foxl2 downregulates SOX9, and targeted disruption of Foxl2 leads to SOX9 upregulation in the XX gonad [130]. Prostaglandin D2 also upregulates SOX9 in the absence of SRY [131].
The role of WNT4 is crucial in the development of the internal genital tract. WNT4 is a secreted protein that functions as a paracrine factor to regulate several developmental mechanisms including the uterus, cervix, and fallopian tubes. In fetuses with XX chromosomes, the absence of SRY releases WNT4 expression, which stabilizes β-catenin and silences SOX9 [132]. β-catenin is responsible for oviduct coiling [133,134]. Many growth factors, such as LIM1, EMX2, HOXA13, PAX2 and 8, and VANGL2 are also essential for the development of reproductive organs. RSPO1 is expressed in the undifferentiated gonadal ridge of XY and XX embryos and increases in the XX gonads in the absence of SRY. RSPO1 binds to G protein-coupled receptors, stimulates the expression of WNT4, and cooperates with it to increase cytoplasmic β-catenin. The increase inWNT4/β-catenin counteracts SOX9, thus leading to the ovarian pathway [135].
There are various genetic mechanisms that are potentially involved in causing a disorder, which can occur in isolation or in combinations to result in a condition. Human genomes are dynamic entities constantly influenced by alterations. The cumulative effects of small-scale sequence alterations (caused by mutation) and larger-scale rearrangements can bring about changes in the genome over a period of time [136]. The genome contains coding regions and non-coding regions. The coding regions of the DNA are directly involved in the formation of proteins, and noncoding regions may or may not be involved in the regulation of gene expression. Regulation of gene expression occurs thanks to long non-coding RNAs and epigenetic, transcriptional, post-transcriptional, translational, and protein location effects [137].
The genes that encode for an important protein related to a particular disease or a physical attribute are called candidate genes [138]. When there are chromosomal deletions, especially of the area encoding for a specific gene, the genetic functions can be completely lost, and resulting phenotypes can be severe [139,140]. Candidate genes for MA are thus thought to be related to the development of the Müllerian or Wolffian duct, or related to regulatory factors like anti-Müllerian hormone (AMH), estrogen, and estrogen receptor. The candidate genes identified by different studies are WNT4, LHX1, HNF1B, and HOXA10 [61,65,70,75,77,116,141,142]. Other genes with possible causative roles not yet substantiated adequately are HNRNPCL1, ITIH5, LRP10, MMRP14, OR2T2, OR4M2, PAX8, PDE11A, RBM8A, SHOX, TBX6, WNT9B, and ZNF816 [Table 1].
A careful balance of levels of different proteins significantly influences the embryonic developmental processes. Duplications of genes encoding these proteins can result in extra gene copies, and the resulting alteration of gene dosing can lead to developmental defects [140]. A proposed mechanism for MA is an overdose of AMH. Duplications were found in many reported studies in multiple locations involving chromosomes 1,2,3,6,7,10,12,16,18,22 and X chromosomes [Table 2]. However, the exact mechanisms of how these changes are related to the causation of MA are yet to be verified.
Mutations involve changes in the nucleotide sequence of a short region of a genome. The number of mutations occurring is usually minimized by the inherent DNA-repair enzymes in the cell, and mutations persist only when the cellular DNA-repair mechanisms fail. The mutations on coding regions are of various types. A lot of mutations are point mutations, where one nucleotide is replaced with the other, or it can involve the insertion or deletion of one or a few nucleotides [136]. Insertions of small numbers of extra nucleotides in the polynucleotide being synthesized, or failure of some nucleotides in the frame being copied, can alter the entire sequence/codon down the frame. Such proteins are usually markedly different from the original proteins. This is called a frame-shift mutation. Silent mutations (also called synchronous) are said to have occurred when changes in the nucleotide sequence have no effect on the functioning of the genome and they do not change the encoded amino acid [165]. Missense mutations are changes that alter a codon to another one, meaning the resultant amino acid is a completely different one. The effects of missense mutations are difficult to predict. If the resultant amino acid is similar to the original one or the change is in a non-critical amino acid, the functions are retained. However, the mutant protein may have a completely different function if the change affects a critical amino acid or the new amino acid is not similar to the original. On the other hand, if the mutation results in the stopping of the translation of the mRNA prematurely because of a stop codon, this is called a nonsense mutation. Nonsense mutation results in a shortened protein, which can be non-functional and this effect depends on how much of the polypeptide is lost [165]. There are various types of mutations documented in the literature, including point mutations, frameshift mutations, and missense mutations in patients with MA [Table 2]. While some of these changes involve candidate genes directly, the significance of others is unclear. It is also noteworthy that DNA methylation levels act as a regulator of gene expression. The presence of DNA methylation, in general, prevents transcriptional activation of genes at a specific cell type [166,167]. A search for altered imprinting markers at the 11p15 imprinting control region 1 (ICR1) in 100 patients with MRKHS failed to detect any defects at that locus [168]. However, it did not rule out the possibility of imprinting alterations at other locations in the etiology of MRKHS.
In a chromosomal translocation, genetic material is exchanged between two chromosomes. Translocations were detected in five different individuals involving chromosomes 8,13, 7,14, and chromosomes 3,16 [40,98,162,163]. These were one individual with t(8;13)(q22.1;q32.1), two with t(8;13)(q12;q14), and one with t(3;16)(p22.3;p13.3). In the latest case report of these by Williams et al., about ten potential genes were identified, and four of significance were further substantiated [66]. As none of the family members in the cases had similar translocations, these were regarded as sporadic occurrences.

3.3. Fertility Options in Women with Müllerian Agenesis

MA means absolute infertility in females, and the diagnosis can be associated with considerable psychological trauma [20]. Hence, the management requires a multidisciplinary approach including gynecologists, fertility specialists, psychologists, clinical nurse specialists, support groups, and counselors [169]. Options for parenthood in these women include surrogacy and uterine transplantation if parents seek a biologically related baby. Adoption is an option if a biologically related baby is not desired in the first place or the options have failed, are unacceptable, or contraindicated [20].
Gestational surrogacy is a popular option in women with AUFI, especially with functioning ovaries, as in MA. The oocytes of the women with MA are collected and fused with the partner’s spermatozoa through in-vitro fertilization. The resulting embryo is then transferred to the uterus of the gestational surrogate. The couple then legally adopts the child from the mother after birth. Legal parenthood of a biologically related child is thus achieved [170]. The woman carrying the pregnancy is called a gestational surrogate. The surrogate can be a close relative of the couple (altruistic) or unrelated, which would be a commercial surrogate. A study by Petrozza et al. did not find evidence of inheritance or congenital anomalies in babies born through surrogacy [96]. However, there are legal implications for this method, which can vary in different countries. Surrogacy as such or specific surrogacy may not be legally permitted in specific countries due to sociocultural issues [20,171,172]. Countries can have different policies regarding commercial or altruistic surrogacy [173,174]. In the United Kingdom, surrogacy is permitted, but surrogacy agreements are not legally enforceable. The surrogate remains the child’s legal mother from birth, up until the parenthood is legally transferred to the intended parents. This is generally carried out after 6 weeks of birth. In case of disputes, the surrogacy arrangement is not legally enforceable [20].
Uterine transplant (UT) involves transplantation of the uterus with the cervix, ligamentous supports, and blood vessels. This results in restoring the natural anatomy. Successful pregnancy and childbirth after a UT ensure conditions akin to natural biological parenthood, which is acceptable both legally and socially. The first live birth following UT was reported in Sweden in 2014 [175]. Since then, numerous case reports and series have been published. A recent review has reported 18 live births out of 45 cases of UT [176]. Typically, the whole process involves procuring a uterus from a live or deceased donor, transplantation, immunosuppression, achieving pregnancy by embryo transfer, and delivery by cesarean section followed by hysterectomy. While live organ donation involves additional risks to the donor due to operative procedures, the clinical outcomes are speculated to be better. Immunosuppression after UT is carried out with a non-teratogenic immunosuppressive regimen and monitored for graft rejection by cervical biopsies [7,177,178]. Embryo transfer is performed using a single euploid blastocyst after 6–12 months [176]. The ensuing pregnancy is then monitored following a protocol for high-risk pregnancy care, and delivered by cesarean section at 37 weeks of gestation, or sooner if indicated. Vaginal delivery is generally not advocated due to concerns about the structural integrity of the graft and sufficiency of vascular anastomoses during contractions. Depending on the reproductive expectations, further embryo transfers can be performed. The uterus is then removed and immunosuppression is stopped to prevent further complications [179]. Although the reproductive scenario in women with MA looks encouraging with UT, it involves ethical issues encompassing both assisted reproduction and organ transplantation [180,181,182].
Adoption is an option for parenthood in couples with MA. It is a legal proceeding that creates a parent–child relationship between persons not related by blood. Adoption laws vary from country to country, and adoption is generally a long process. While it can be legally and socially acceptable, the child is not biologically related to the parents.

4. Conclusions

MA results in absolute uterine factor infertility. The genetic basis of MA is yet unclear and etiology is mostly multifactorial. Although many candidate genes have been identified, more studies are required to substantiate the evidence. With the advancing options for parenthood of biologically related offspring, further studies will be possible to identify candidate genes, accurate mechanisms of MA, and inheritance of this condition.

Supplementary Materials

The supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25010120/s1.

Author Contributions

Conceptualization, S.S.K., M.J., B.T.G. and R.D.; methodology, R.D. and S.S.K.; software, M.J. and B.T.G.; validation, R.D. and S.S.K.; formal analysis, M.J. and R.D.; investigation, S.S.K., M.J., B.T.G. and R.D.; resources, B.T.G. and S.S.K.; data curation, R.D. and S.S.K.; writing—original draft preparation, R.D. and S.S.K.; writing—review and editing, M.J. and B.T.G.; visualization, S.S.K. and R.D.; supervision, M.J., B.T.G., S.S.K. and R.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization (WHO). International Classification of Diseases; 11th Revision (ICD-11); WHO: Geneva, Switzerland, 2018. [Google Scholar]
  2. Larsen, U. Research on infertility: Which definition should we use? Fertil. Steril. 2005, 83, 846–852. [Google Scholar] [CrossRef] [PubMed]
  3. World Health Organization. WHO fact sheet on infertility. Glob. Reprod. Health 2021, 6, e52. [Google Scholar] [CrossRef]
  4. Assistance Médicale à la Procréation (AMP). Inserm-La Science Pour la Santé n.d. Available online: https://www.inserm.fr/information-en-sante/dossiers-information/assistance-medicale-procreation-amp (accessed on 7 August 2022).
  5. Dube, R.; Kar, S.S. Genital Microbiota and Outcome of Assisted Reproductive Treatment—A Systematic Review. Life 2022, 12, 1867. [Google Scholar] [CrossRef] [PubMed]
  6. Sallée, C.; Margueritte, F.; Marquet, P.; Piver, P.; Aubard, Y.; Lavoué, V.; Dion, L.; Gauthier, T. Uterine Factor Infertility, a Sys-tematic Review. J. Clin. Med. 2022, 11, 4907. [Google Scholar] [CrossRef] [PubMed]
  7. Brännström, M.; Johannesson, L.; Dahm-Kähler, P.; Enskog, A.; Mölne, J.; Kvarnström, N.; Diaz-Garcia, C.; Hanafy, A.; Lundmark, C.; Marcickiewicz, J.; et al. First clinical uterus transplantation trial: A six-month report. Fertil. Steril. 2014, 101, 1228–1236. [Google Scholar] [CrossRef] [PubMed]
  8. Hur, C.; Rehmer, J.; Flyckt, R.; Falcone, T. Uterine Factor Infertility: A Clinical Review. Clin. Obstet. Gynecol. 2019, 62, 257–270. [Google Scholar] [CrossRef] [PubMed]
  9. Moncada-Madrazo, M.; Rodríguez Valero, C. Embryology, Uterus; Updated 25 July 2023; StatPearls: Treasure Island, FL, USA, 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK547748/ (accessed on 30 August 2023).
  10. Robbins, J.B.; Broadwell, C.; Chow, L.C.; Parry, J.P.; Sadowski, E.A. Müllerian duct anomalies: Embryological development, classification, and MRI assessment. J. Magn. Reson. Imaging 2015, 41, 1–12. [Google Scholar] [CrossRef] [PubMed]
  11. Ghofrani, M. Embryology. PathologyOutlines.com Website. Available online: https://www.pathologyoutlines.com/topic/ovarynontumorembryology.html (accessed on 28 September 2023).
  12. Dube, R.; Al-Zuheiri, S.T.S.; Syed, M.; Harilal, L.; Zuhaira, D.A.L.; Kar, S.S. Prevalence, Clinico-Bacteriological Profile, and Antibiotic Resistance of Symptomatic Urinary Tract Infections in Pregnant Women. Antibiotics 2023, 12, 33. [Google Scholar] [CrossRef]
  13. Dube, R.; Kar, S.S. COVID-19 in pregnancy: The foetal perspective—A systematic review. BMJ Paediatr. Open 2020, 4, e000859. [Google Scholar] [CrossRef]
  14. AlZuheiri, S.T.; Dube, R.; Menezes, G.; Qasem, S. Clinical profile and outcome of Group B streptococcal colonization in moth-ers and neonates in Ras Al Khaimah, United Arab Emirates: A prospective observational study. Saudi J. Med. Med. Sci. 2021, 9, 235–240. [Google Scholar]
  15. Fontana, L.; Gentilin, B.; Fedele, L.; Gervasini, C.; Miozzo, M. Genetics of Mayer-Rokitansky-Küster-Hauser (MRKH) syn-drome. Clin. Genet. 2017, 91, 233–246. [Google Scholar] [CrossRef] [PubMed]
  16. Morcel, K.; Camborieux, L.; Programme de Recherches sur les Aplasies Müllériennes (PRAM); Guerrier, D. May-er-Rokitansky-Küster-Hauser (MRKH) syndrome. Orphanet J. Rare Dis. 2007, 14, 13. [Google Scholar] [CrossRef] [PubMed]
  17. Oppelt, P.; Strissel, P.; Kellermann, A.; Seeber, S.; Humeny, A.; Beckmann, M.; Strick, R. DNA sequence variations of the entire anti-Müllerian hormone (AMH) gene promoter and AMH protein expression in patients with the Mayer–Rokitanski–Küster–Hauser syndrome. Hum. Reprod. 2005, 20, 149–157. [Google Scholar] [CrossRef] [PubMed]
  18. Guerrier, D.; Mouchel, T.; Pasquier, L.; Pellerin, I. The Mayer-Rokitansky-Küster-Hauser syndrome (congenital absence of uterus and vagina)—Phenotypic manifestations and genetic approaches. J. Negat. Results Biomed. 2006, 5, 1. [Google Scholar] [CrossRef] [PubMed]
  19. Herlin, M.; Højland, A.T.; Petersen, M.B. Familial occurrence of Mayer-Rokitansky-Küster-Hauser syndrome: A case report and review of the literature. Am. J. Med. Genet. A 2014, 164A, 2276–2286. [Google Scholar] [CrossRef]
  20. Jones, B.P.; Ranaei-Zamani, N.; Vali, S.; Williams, N.; Saso, S.; Thum, M.Y.; Al-Memar, M.; Dixon, N.; Rose, G.; Testa, G.; et al. Options for acquiring motherhood in abso-lute uterine factor infertility; adoption, surrogacy and uterine transplantation. Obstet. Gynaecol. 2021, 23, 138–147. [Google Scholar] [CrossRef] [PubMed]
  21. Ledig, S.; Schippert, C.; Strick, R.; Beckmann, M.W.; Oppelt, P.G.; Wieacker, P. Recurrent aberrations identified by array-CGH in patients with Mayer-Rokitansky-Küster-Hauser syndrome. Fertil. Steril. 2011, 95, 1589–1594. [Google Scholar] [CrossRef]
  22. Ma, D.; Marion, R.; Punjabi, N.P.; Pereira, E.; Samanich, J.; Agarwal, C.; Li, J.; Huang, C.-K.; Ramesh, K.H.; Cannizzaro, A.L.; et al. A de novo 10.79 Mb interstitial deletion at 2q13q14.2 involving PAX8 causing hypothyroidism and mullerian agenesis: A novel case report and literature review. Mol. Cytogenet. 2014, 7, 85. [Google Scholar] [CrossRef]
  23. Chen, M.-J.; Wei, S.-Y.; Yang, W.-S.; Wu, T.-T.; Li, H.-Y.; Ho, H.-N.; Yang, Y.-S.; Chen, P.-L. Concurrent exome-targeted next generation sequencing and single nucleotide polymorphism array to identify the causative genetic aberrations of isolated Mayer-Rokitansky-Kuster-Hauser syndrome. Hum. Reprod. 2015, 30, 1732–1742. [Google Scholar] [CrossRef]
  24. Lischke, J.H.; Curtis, C.H.; Lamb, E.J. Discordance of vaginal agenesis in monozygotic twins. Obstet. Gynecol. 1973, 41, 920–924. [Google Scholar]
  25. Rall, K.; Eisenbeis, S.; Barresi, G.; Rückner, D.; Walter, M.; Poths, S.; Wallwiener, D.; Riess, O.; Bonin, M.; Brucker, S. May-er-Rokitansky-Küster-Hauser syndrome discordance in monozygotic twins: Matrix metalloproteinase 14, low-density lip-oprotein receptor–related protein 10, extracellular matrix, and neoangiogenesis genes identified as candidate genes in a tissue-specific mosaicism. Fertil. Steril. 2015, 103, 494–502.e3. [Google Scholar] [PubMed]
  26. Heidenreich, W.; Pfeiffer, A.; Kumbnani, H.K.; Scholz, W.; Zeuner, W. Disordant monozygotic twins with Mayer Rokitansky Kütser syndrome (author’s transl). Geburtshilfe Und Frauenheilkd. 1977, 37, 221–223. [Google Scholar]
  27. Steinkampf, M.P.; Dharia, S.P.; Dickerson, R.D. Monozygotic twins discordant for vaginal agenesis and bilateral tibial longi-tudinal deficiency. Fertil. Steril. 2003, 80, 643–645. [Google Scholar] [CrossRef] [PubMed]
  28. Regenstein, A.C.; Berkeley, A.S. Discordance of müllerian agenesis in monozygotic twins. A case report. J. Reprod. Med. 1991, 36, 396–397. [Google Scholar] [PubMed]
  29. Duru, U.A.; Laufer, M.R. Discordance in Mayer-von Rokitansky-Küster-Hauser Syndrome Noted in Monozygotic Twins. J. Pediatr. Adolesc. Gynecol. 2009, 22, e73–e75. [Google Scholar] [CrossRef] [PubMed]
  30. Milsom, S.R.; Ogilvie, C.M.; Jefferies, C.; Cree, L. Discordant Mayer-Rokitansky-Kuster-Hauser (MRKH) syndrome in identi-cal twins—A case report and implications for reproduction in MRKH women. Gynecol. Endocrinol. 2015, 31, 684–687. [Google Scholar] [CrossRef] [PubMed]
  31. Maniglio, P.; Ricciardi, E.; Laganà, A.S.; Triolo, O.; Caserta, D. Epigenetic modifications of primordial reproductive tract: A common etiologic pathway for Mayer-Rokitansky-Kuster-Hauser Syndrome and endometriosis? Med. Hypotheses 2016, 90, 4–5. [Google Scholar] [CrossRef]
  32. Rall, K.; Barresi, G.; Walter, M.; Poths, S.; Haebig, K.; Schaeferhoff, K.; Schoenfisch, B.; Riess, O.; Wallwiener, D.; Bonin, M.; et al. A combination of transcriptome and methylation analyses reveals embryologically-relevant candidate genes in MRKH patients. Orphanet J. Rare Dis. 2011, 6, 32. [Google Scholar] [CrossRef]
  33. Ray, R.; Kar, S.S.; Mahapatro, S.; Tripathy, R.; Mohanty, R. A Rare cause of seizure-dysgenesis of corpus callosum. Indian J. Pract. Pediatr. 2009, 11, 101–104. [Google Scholar]
  34. Theisen, A. Microarray-based Comparative Genomic Hybridization (aCGH). Nat. Educ. 2008, 1, 45. [Google Scholar]
  35. Eleanor, G.; Seaby, R.J.; Pengelly, S.E. Exome sequencing explained: A practical guide to its clinical application. Brief. Funct. Genom. 2016, 15, 374–384. [Google Scholar] [CrossRef]
  36. Van Dijk, E.L.; Jaszczyszyn, Y.; Naquin, D.; Thermes, C. The Third Revolution in Sequencing Technology. Trends Genet. 2018, 34, 666–681. [Google Scholar] [CrossRef] [PubMed]
  37. Sahyouni, J.K.; Odeh, L.B.M.; Mulla, F.; Junaid, S.; Kar, S.S.; Almarri, N.M.J.A.B. Infantile Sandhoff disease with ventricular septal defect: A case report. J. Med. Case Rep. 2022, 16, 317. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, N.; Zhao, S.; Jolly, A.; Wang, L.; Pan, H.; Yuan, J.; Chen, S.; Koch, A.; Ma, C.; Tian, W.; et al. Perturbations of genes essential for Mullerian duct and Wolffian duct development in Mayer-Rokitansky-Kuster-Hauser syndrome. Am. J. Hum. Genet. 2021, 108, 337–345. [Google Scholar] [CrossRef] [PubMed]
  39. Mikhael, S.; Dugar, S.; Morton, M.; Chorich, L.P.; Tam, K.B.; Lossie, A.C.; Kim, H.-G.; Knight, J.; Taylor, H.S.; Mukherjee, S.; et al. Genetics of agenesis/hypoplasia of the uterus and vagina: Narrowing down the number of candidate genes for Mayer–Rokitansky–Küster–Hauser Syndrome. Qual. Life Res. 2021, 140, 667–680. [Google Scholar] [CrossRef]
  40. Williams, L.S.; Kim, H.G.; Kalscheuer, V.M.; Tuck, J.M.; Chorich, L.P.; Sullivan, M.E.; Falkenstrom, A.; Reindollar, R.H.; Layman, L.C. A balanced chromosomal translocation involving chromosomes 3 and 16 in a patient with Mayer-Rokitansky-Kuster-Hauser syndrome reveals new candidate genes at 3p22.3 and 16p13.3. Mol. Cytogenet. 2016, 9, 57. [Google Scholar] [CrossRef]
  41. Pan, H.-X.; Luo, G.-N.; Wan, S.-Q.; Qin, C.-L.; Tang, J.; Zhang, M.; Du, M.; Xu, K.-K.; Shi, J.-Q. Detection of de novo genetic var-iants in Mayer–Rokitansky–Küster–Hauser syndrome by whole genome sequencing. Eur. J. Obstet. Gynecol. Reprod. Biol. X 2019, 4, 100089. [Google Scholar]
  42. Thomson, E.; Tran, M.; Robevska, G.; Ayers, K.; van der Bergen, J.; Bhaskaran, P.G.; Haan, E.; Cereghini, S.; Vash-Margita, A.; Margetts, M.; et al. Functional genomics analysis identifies loss of HNF1B function as a cause of Mayer-Rokitansky-Küster-Hauser syndrome. Hum. Mol. Genet. 2023, 32, 1032–1047. [Google Scholar] [CrossRef]
  43. Backhouse, B.; Hanna, C.; Robevska, G.; Bergen, J.V.D.; Pelosi, E.; Simons, C.; Koopman, P.; Juniarto, A.Z.; Grover, S.; Faradz, S.; et al. Identification of Candidate Genes for Mayer-Rokitansky-Küster-Hauser Syndrome Using Genomic Approaches. Sex. Dev. 2019, 13, 26–34. [Google Scholar] [CrossRef]
  44. Chu, C.; Li, L.; Li, S.; Zhou, Q.; Zheng, P.; Zhang, Y.-D.; Duan, A.-H.; Lu, D.; Wu, Y.-M. Variants in genes related to develop-ment of the urinary system are associated with Mayer–Rokitansky–Küster–Hauser syndrome. Hum. Genom. 2022, 16, 10. [Google Scholar] [CrossRef]
  45. Pontecorvi, P.; Bernardini, L.; Capalbo, A.; Ceccarelli, S.; Megiorni, F.; Vescarelli, E.; Bottillo, I.; Preziosi, N.; Fabbretti, M.; Perniola, G.; et al. Protein–protein interaction network analysis applied to DNA copy number profiling suggests new per-spectives on the aetiology of Mayer–Rokitansky–Küster–Hauser syndrome. Sci. Rep. 2021, 11, 448. [Google Scholar] [CrossRef] [PubMed]
  46. Nodale, C.; Ceccarelli, S.; Giuliano, M.; Cammarota, M.; D’Amici, S.; Vescarelli, E.; Maffucci, D.; Bellati, F.; Panici, P.B.; Ro-mano, F.; et al. Gene Expression Profile of Patients with Mayer-Rokitansky-Küster-Hauser Syndrome: New Insights into the Potential Role of Developmental Pathways. PLoS ONE 2014, 9, e91010. [Google Scholar] [CrossRef] [PubMed]
  47. Cheroki, C.; Krepischi, A.; Szuhai, K.; Brenner, V.; Kim, C.; Otto, P.A.; Rosenberg, C. Genomic imbalances associated with mullerian aplasia. J. Med. Genet. 2007, 45, 228–232. [Google Scholar] [CrossRef] [PubMed]
  48. McGowan, R.; Tydeman, G.; Shapiro, D.; Craig, T.; Morrison, N.; Logan, S.; Balen, A.H.; Ahmed, S.F.; Deeny, M.; Tolmie, J.; et al. DNA copy number variations are important in the complex genetic architecture of müllerian disorders. Fertil. Steril. 2015, 103, 1021–1030.e1. [Google Scholar] [CrossRef] [PubMed]
  49. Ledig, S.; Wieacker, P. Clinical and genetic aspects of Mayer–Rokitansky–Küster–Hauser syndrome. Med. Genet. 2018, 30, 3–11. [Google Scholar] [CrossRef] [PubMed]
  50. Tewes, A.-C.; Rall, K.K.; Römer, T.; Hucke, J.; Kapczuk, K.; Brucker, S.; Wieacker, P.; Ledig, S. Variations in RBM8A and TBX6 are associated with disorders of the müllerian ducts. Fertil. Steril. 2015, 103, 1313–1318. [Google Scholar] [CrossRef] [PubMed]
  51. Ma, W.; Li, Y.; Wang, M.; Li, H.; Su, T.; Wang, S. Associations of Polymorphisms in WNT9B and PBX1 with May-er-Rokitansky-Küster-Hauser Syndrome in Chinese Han. PLoS ONE 2015, 10, e0130202. [Google Scholar]
  52. Wang, M.; Li, Y.; Ma, W.; Li, H.; He, F.; Pu, D.; Su, T.; Wang, S. Analysis of WNT9B mutations in Chinese women with Mayer–Rokitansky–Küster–Hauser syndrome. Reprod. Biomed. Online 2014, 28, 80–85. [Google Scholar] [CrossRef]
  53. Buchert, R.; Schenk, E.; Hentrich, T.; Weber, N.; Rall, K.; Sturm, M.; Kohlbacher, O.; Koch, A.; Riess, O.; Brucker, S.Y.; et al. Ge-nome Sequencing and Transcriptome Profiling in Twins Discordant for Mayer-Rokitansky-Küster-Hauser Syndrome. J. Clin. Med. 2022, 11, 5598. [Google Scholar] [CrossRef]
  54. Waschk, D.; Tewes, A.-C.; Römer, T.; Hucke, J.; Kapczuk, K.; Schippert, C.; Hillemanns, P.; Wieacker, P.; Ledig, S. Mutations in WNT9B are associated with Mayer-Rokitansky-Küster-Hauser syndrome. Clin. Genet. 2016, 89, 590–596. [Google Scholar] [CrossRef]
  55. Nik-Zainal, S.; Strick, R.; Storer, M.; Huang, N.; Rad, R.; Willatt, L.; Fitzgerald, T.; Martin, V.; Sandford, R.; Carter, N.P.; et al. High incidence of recurrent copy number variants in patients with isolated and syndromic Mullerian aplasia. J. Med. Genet. 2011, 48, 197–204. [Google Scholar] [CrossRef] [PubMed]
  56. Sandbacka, M.; Laivuori, H.; Freitas, É.; Halttunen, M.; Jokimaa, V.; Morin-Papunen, L.; Rosenberg, C.; Aittomäki, K. TBX6, LHX1 and copy number variations in the complex genetics of Müllerian aplasia. Orphanet J. Rare Dis. 2013, 8, 125. [Google Scholar] [CrossRef] [PubMed]
  57. Eksi, D.D.; Shen, Y.; Erman, M.; Chorich, L.P.; Sullivan, M.E.; Bilekdemir, M.; Yılmaz, E.; Luleci, G.; Kim, H.-G.; Alper, O.M.; et al. Copy number variation and regions of homozygosity analysis in patients with MÜLLERIAN aplasia. Mol. Cytogenet. 2018, 11, 13. [Google Scholar] [CrossRef] [PubMed]
  58. Tewes, A.-C.; Hucke, J.; Römer, T.; Kapczuk, K.; Schippert, C.; Hillemanns, P.; Wieacker, P.; Ledig, S. Sequence Variants in TBX6 Are Associated with Disorders of the Müllerian Ducts: An Update. Sex. Dev. 2019, 13, 35–40. [Google Scholar] [CrossRef] [PubMed]
  59. Ma, C.; Chen, N.; Jolly, A.; Zhao, S.; Coban-Akdemir, Z.; Tian, W.; Kang, J.; Ye, Y.; Wang, Y.; Koch, A.; et al. Functional characteristics of a broad spectrum of TBX6 variants in Mayer-Rokitansky-Küster-Hauser syndrome. Genet. Med. Off. J. Am. Coll. Med. Genet. 2022, 24, 2262–2273. [Google Scholar] [CrossRef] [PubMed]
  60. Bernardini, L.; Gimelli, S.; Gervasini, C.; Carella, M.; Baban, A.; Frontino, G.; Barbano, G.; Divizia, M.T.; Fedele, L.; Novelli, A.; et al. Recurrent microdeletion at 17q12 as a cause of Mayer-Rokitansky-Kuster-Hauser (MRKH) syndrome: Two case reports. Orphanet J. Rare Dis. 2009, 4, 25. [Google Scholar] [CrossRef] [PubMed]
  61. Ledig, S.; Brucker, S.; Barresi, G.; Schomburg, J.; Rall, K.; Wieacker, P. Frame shift mutation of LHX1 is associated with May-er-Rokitansky-Kuster-Hauser (MRKH) syndrome. Hum. Reprod. 2012, 27, 2872–2875. [Google Scholar] [CrossRef]
  62. Hinkes, B.; Hilgers, K.F.; Bolz, H.J.; Goppelt-Struebe, M.; Amann, K.; Nagl, S.; Bergmann, C.; Rascher, W.; Eckardt, K.-U.; Jaco-bi, J. A complex microdeletion 17q12 phenotype in a patient with recurrent de novo membranous nephropathy. BMC Nephrol. 2012, 13, 27. [Google Scholar] [CrossRef]
  63. Ledig, S.; Tewes, A.; Hucke, J.; Römer, T.; Kapczuk, K.; Schippert, C.; Hillemanns, P.; Wieacker, P. Array-comparative genomic hybridization analysis in patients with Müllerian fusion anomalies. Clin. Genet. 2018, 93, 640–646. [Google Scholar] [CrossRef]
  64. Mencarelli, M.A.; Katzaki, E.; Papa, F.T.; Sampieri, K.; Caselli, R.; Uliana, V.; Pollazzon, M.; Canitano, R.; Mostardini, R.; Grosso, S.; et al. Private inherited microdeletion/microduplications: Implications in clinical practice. Eur. J. Med. Genet. 2008, 51, 409–416. [Google Scholar] [CrossRef]
  65. Lindner, T.H.; Njølstad, P.R.; Horikawa, Y.; Bostad, L.; Bell, G.I.; Søvik, O. A novel syndrome of diabetes mellitus, renal dys-function and genital malformation associated with a partial deletion of the pseudo-POU domain of hepatocyte nuclear fac-tor-1beta. Hum. Mol. Genet. 1999, 8, 2001–2008. [Google Scholar] [CrossRef] [PubMed]
  66. Williams, L.S.; Eksi, D.D.; Shen, Y.; Lossie, A.C.; Chorich, L.P.; Sullivan, M.E.; Phillips, J.A.; Erman, M.; Kim, H.-G.; Alper, O.M.; et al. Genetic analysis of Mayer-Rokitansky-Kuster-Hauser syndrome in a large cohort of families. Fertil. Steril. 2017, 108, 145–151.e2. [Google Scholar] [CrossRef] [PubMed]
  67. Xia, M.; Zhao, H.; Qin, Y.; Mu, Y.; Wang, J.; Bian, Y.; Ma, J.; Chen, Z.-J. LHX1 mutation screening in 96 patients with müllerian duct abnormalities. Fertil. Steril. 2012, 97, 682–685. [Google Scholar] [CrossRef] [PubMed]
  68. Zhang, W.; Zhou, X.; Liu, L.; Zhu, Y.; Liu, C.; Pan, H. Identification and functional analysis of a novel LHX1 mutation associ-ated with congenital absence of the uterus and vagina. Oncotarget 2017, 8, 8785–8790. [Google Scholar] [CrossRef] [PubMed]
  69. Oram, R.A.; Edghill, E.L.; Blackman, J.; Taylor, M.J.; Kay, T.; Flanagan, S.E.; Ismail-Pratt, I.; Creighton, S.M.; Ellard, S.; Hatters-ley, A.T.; et al. Mutations in the hepatocyte nuclear factor-1_ (HNF1B) gene are common with combined uterine and renal malformations but are not found with isolated uterine malformations. Am. J. Obstet. Gynecol. 2010, 203, 364.e1–364.e5. [Google Scholar] [CrossRef] [PubMed]
  70. Bingham, C.; Ellard, S.; Cole, T.R.; Jones, K.E.; Allen, L.I.; Goodship, J.A.; Goodship, T.H.; Bakalinova-Pugh, D.; Russell, G.I.; Woolf, A.; et al. Solitary functioning kidney and diverse genital tract malformations associated with hepatocyte nuclear factor-1 mutations. Kidney Int. 2002, 61, 1243–1251. [Google Scholar] [CrossRef] [PubMed]
  71. Sundaram, U.T.; McDonald-McGinn, D.M.; Huff, D.; Emanuel, B.S.; Zackai, E.H.; Driscoll, D.A.; Bodurtha, J. Primary amenor-rhea and absent uterus in the 22q11.2 deletion syndrome. Am. J. Med. Genet. Part A 2007, 143, 2016–2018. [Google Scholar] [CrossRef]
  72. Morcel, K.; Watrin, T.; Pasquier, L.; Rochard, L.; Le Caignec, C.; Dubourg, C.; Loget, P.; Paniel, B.-J.; Odent, S.; David, V.; et al. Utero-vaginal aplasia (Mayer-Rokitansky-Küster-Hauser syndrome) associated with deletions in known DiGeorge or Di-George-like loci. Orphanet J. Rare Dis. 2011, 6, 9. [Google Scholar] [CrossRef]
  73. Cheroki, C.; Krepischi-Santos, A.C.; Rosenberg, C.; Jehee, F.S.; Mingroni-Netto, R.C.; Filho, I.P.; Filho, S.Z.; Kim, C.A.; Bagnoli, V.R.; Mendonca, B.B.; et al. Report of a del22q11 in a patient with Mayer-Rokitansky-Küster-Hauser (MRKH) anomaly and exclusion ofWNT-4,RAR-gamma, andRXR-alpha as major genes determining MRKH anomaly in a study of 25 affected women. Am. J. Med. Genet. Part A 2006, 140, 1339–1342. [Google Scholar] [CrossRef]
  74. AlSubaihin, A.; Vandermeulen, J.; Harris, K.; Duck, J.; McCready, E. Müllerian Agenesis in Cat Eye Syndrome and 22q11 Chromosome Abnormalities: A Case Report and Literature Review. J. Pediatr. Adolesc. Gynecol. 2018, 31, 158–161. [Google Scholar] [CrossRef]
  75. Philibert, P.; Biason-Lauber, A.; Gueorguieva, I.; Stuckens, C.; Pienkowski, C.; Lebon-Labich, B.; Paris, F.; Sultan, C. Molecular analysis of WNT4 gene in four adolescent girls with mullerian duct abnormality and hyperandrogenism (atypical Mayer-Rokitansky-Küster-Hauser syndrome). Fertil. Steril. 2011, 95, 2683–2686. [Google Scholar] [CrossRef] [PubMed]
  76. Ragitha, T.S.; Sunish, K.S.; Gilvaz, S.; Daniel, S.; Varghese, P.R.; Raj, S.; Francis, J.; Kumar, R.S. Mutation analysis of WNT4 gene in SRY negative 46,XX DSD patients with Mullerian agenesis and/or gonadal dysgenesis-An Indian study. Gene 2023, 861, 147236. [Google Scholar] [CrossRef] [PubMed]
  77. Biason-Lauber, A.; Konrad, D.; Navratil, F.; Schoenle, E.J. A WNT4 Mutation Associated with Müllerian-Duct Regression and Virilization in a 46,XXWoman. N. Engl. J. Med. 2004, 351, 792–798. [Google Scholar] [CrossRef] [PubMed]
  78. Kyei Barffour, I.; Kyei Baah Kwarkoh, R. GREB1L as a candidate gene of Mayer-Rokitansky-Kuster-Hauser Syndrome. Eur. J. Med. Genet. 2021, 64, 104158. [Google Scholar] [CrossRef] [PubMed]
  79. Herlin, M.K.; Le-Quy, V.; Højland, A.T.; Ernst, A.; Okkels, H.; Petersen, A.C.; Petersen, M.B.; Pedersen, I.S. Whole-exome se-quencing identifies a GREB1L variant in a three-generation family with Müllerian and renal agenesis: A novel candidate gene in Mayer–Rokitansky–Küster–Hauser (MRKH) syndrome. A case report. Hum. Reprod. 2019, 34, 1838–1846. [Google Scholar] [CrossRef] [PubMed]
  80. Jacquinet, A.; Boujemla, B.; Fasquelle, C.; Thiry, J.; Josse, C.; Lumaka, A.; Brischoux-Boucher, E.; Dubourg, C.; David, V.; Pas-quier, L.; et al. GREB1L variants in familial and sporadic hereditary urogenital adysplasia and May-er-Rokitansky-Kuster-Hauser syndrome. Clin. Genet. 2020, 98, 126–137. [Google Scholar] [CrossRef]
  81. Xing, Q.; Xu, Z.; Zhu, Y.; Wang, X.; Wang, J.; Chen, D.; Xu, Y.; He, X.; Xiang, H.; Wang, B.; et al. Genetic analysis of DACT1 in 100 Chinese Han women with Müllerian duct anomalies. Reprod. Biomed. Online 2016, 32, 420–426. [Google Scholar] [CrossRef]
  82. Ravel, C.; Bashamboo, A.; Bignon-Topalovic, J.; Siffroi, J.-P.; McElreavey, K.; Darai, E. Polymorphisms in DLGH1 and LAMC1 in Mayer–Rokitansky–Kuster–Hauser syndrome. Reprod. Biomed. Online 2012, 24, 462–465. [Google Scholar] [CrossRef]
  83. Brucker, S.Y.; Frank, L.; Eisenbeis, S.; Henes, M.; Wallwiener, D.; Riess, O.; van Eijck, B.; Schöller, D.; Bonin, M.; Rall, K.K. Se-quence variants in ESR1 and OXTR are associated with Mayer-Rokitansky-Küster-Hauser syndrome. Acta Obstet. Gynecol. Scand. 2017, 96, 1338–1346. [Google Scholar] [CrossRef]
  84. Brucker, S.Y.; Eisenbeis, S.; Konig, J.; Lamy, M.; Salker, M.S.; Zeng, N.; Seeger, H.; Henes, M.; Scholler, D.; Schonfisch, B.; et al. Decidualization is Impaired in Endometrial Stromal Cells from Uterine Rudiments in Mayer-Rokitansky-Kuster-Hauser Syndrome. Cell Physiol. Biochem. 2017, 41, 1083–1097. [Google Scholar] [CrossRef]
  85. Miyamoto, Y.; Taniguchi, H.; Hamel, F.; Silversides, D.W.; Viger, R.S. A GATA4/WT1 cooperation regulates transcription of genes required for mammalian sex determination and differentiation. BMC Mol Biol. 2008, 9, 44. [Google Scholar] [CrossRef] [PubMed]
  86. Li, H.; Liao, S.; Luo, G.; Li, H.; Wang, S.; Li, Z.; Luo, X. An Association between EMX2 Variations and May-er-Rokitansky-Küster-Hauser Syndrome: A Case-Control Study of Chinese Women. J. Healthc. Eng. 2022, 2022, 9975369. [Google Scholar] [PubMed]
  87. Liu, S.; Gao, X.; Qin, Y.; Liu, W.; Huang, T.; Ma, J.; Simpson, J.L.; Chen, Z.-J. Nonsense mutation of EMX2 is potential causative for uterus didelphysis: First molecular explanation for isolated incomplete müllerian fusion. Fertil. Steril. 2015, 103, 769–774.e2. [Google Scholar] [CrossRef] [PubMed]
  88. Smol, T.; Ribero-Karrouz, W.; Edery, P.; Gorduza, D.B.; Catteau-Jonard, S.; Manouvrier-Hanu, S.; Ghoumid, J. May-er-Rokitansky-Kunster-Hauser syndrome due to 2q12.1q14.1 deletion: PAX8 the causing gene? Eur. J. Med. Genet. 2020, 63, 103812. [Google Scholar] [CrossRef]
  89. Gervasini, C.; Grati, F.R.; Lalatta, F.; Tabano, S.; Gentilin, B.; Colapietro, P.; De Toffol, S.; Frontino, G.; Motta, F.; Maitz, S.; et al. SHOX duplications found in some cases with type I Mayer-Rokitansky-Kuster-Hauser syndrome. Genet. Med. 2010, 12, 634–640. [Google Scholar] [CrossRef]
  90. Ma, J.; Qin, Y.; Liu, W.; Duan, H.; Xia, M.; Chen, Z.-J. Analysis of PBX1 mutations in 192 Chinese women with Müllerian duct abnormalities. Fertil. Steril. 2011, 95, 2615–2617. [Google Scholar] [CrossRef]
  91. National Center for Biotechnology Information. Available online: https://www.ncbi.nlm.nih.gov/gene/7849 (accessed on 10 September 2023).
  92. Timmreck, L.S.; Gray, M.R.; Handelin, B.; Allito, B.; Rohlfs, E.; Davis, A.J.; Gidwani, G.; Reindollar, R.H. Analysis of cystic fi-brosis transmembrane conductance regulator gene mutations in patients with congenital absence of the uterus and vagina. Am. J. Med. Genet. 2003, 120, 72–76. [Google Scholar] [CrossRef]
  93. Drummond, J.B.; Rezende, C.F.; Peixoto, F.C.; Carvalho, J.S.; Reis, F.M.; De Marco, L. Molecular analysis of the beta-catenin gene in patients with the Mayer-Rokitansky-Küster-Hauser syndrome. J. Assist. Reprod. Genet. 2008, 25, 511–514. [Google Scholar] [CrossRef]
  94. Lalwani, S.; Wu, H.-H.; Reindollar, R.H.; Gray, M.R. HOXA10 mutations in congenital absence of uterus and vagina. Fertil. Steril. 2008, 89, 325–330. [Google Scholar] [CrossRef]
  95. Takahashi, K.; Hayano, T.; Sugimoto, R.; Kashiwagi, H.; Shinoda, M.; Nishijima, Y.; Suzuki, T.; Suzuki, S.; Ohnuki, Y.; Kondo, A.; et al. Exome and copy number variation analyses of Mayer–Rokitansky–Küster–Hauser syndrome. Hum. Genome Var. 2018, 5, 27. [Google Scholar] [CrossRef]
  96. Petrozza, J.C.; Gray, M.R.; Davis, A.J.; Reindollar, R.H. Congenital absence of the uterus and vagina is not commonly transmitted as a dominant genetic trait: Outcomes of surrogate pregnancies. Fertil. Steril. 1997, 67, 387–389. [Google Scholar] [CrossRef] [PubMed]
  97. Hentrich, T.; Koch, A.; Weber, N.; Kilzheimer, A.; Maia, A.; Burkhardt, S.; Rall, K.; Casadei, N.; Kohlbacher, O.; Riess, O.; et al. The Endometrial Transcription Landscape of MRKH Syndrome. Front. Cell Dev. Biol. 2020, 8, 572281. [Google Scholar] [CrossRef] [PubMed]
  98. Brakta, S.; Hawkins, Z.A.; Sahajpal, N.; Seman, N.; Kira, D.; Chorich, L.P.; Kim, H.G.; Xu, H.; Phillips, J.A., 3rd; Kolhe, R.; et al. Rare structural variants, aneuploidies, and mosaicism in in-dividuals with Mullerian aplasia detected by optical genome mapping. Hum Genet. 2023, 142, 483–494. [Google Scholar] [CrossRef] [PubMed]
  99. Murry, J.B.; Santos, X.M.; Wang, X.; Wan, Y.-W.; Veyver, I.B.V.D.; Dietrich, J.E. A genome-wide screen for copy number altera-tions in an adolescent pilot cohort with müllerian anomalies. Fertil. Steril. 2015, 103, 487–493. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, P.; Zhao, H.; Sun, M.; Li, Y.; Chen, Z.-J. PAX2 in 192 Chinese women with Müllerian duct abnormalities: Mutation analysis. Reprod. Biomed. Online 2012, 25, 219–222. [Google Scholar] [CrossRef] [PubMed]
  101. Chang, X.; Qin, Y.; Xu, C.; Li, G.; Zhao, X.; Chen, Z.-J. Mutations in WNT4 are not responsible for Müllerian duct abnormali-ties in Chinese women. Reprod. Biomed. Online 2012, 24, 630–633. [Google Scholar] [CrossRef] [PubMed]
  102. Liatsikos, S.A.; Grimbizis, G.F.; Georgiou, I.; Papadopoulos, N.; Lazaros, L.; Bontis, J.N.; Tarlatzis, B.C. HOX A10 and HOX A11 mutation scan in congenital malformations of the female genital tract. Reprod. Biomed. Online 2010, 21, 126–132. [Google Scholar] [CrossRef]
  103. Hofstetter, G.; Concin, N.; Marth, C.; Rinne, T.; Erdel, M.; Janecke, A. Genetic analyses in a variant of May-er-Rokitansky-Kuster-Hauser syndrome (MURCS association). Wien. Klin. Wochenschr. 2008, 120, 435–439. [Google Scholar] [CrossRef]
  104. Burel, A.; Mouchel, T.; Odent, S.; Tiker, F.; Knebelmann, B.; Pellerin, I.; Guerrier, D. Role of HOXA7 to HOXA13 and PBX1 genes in various forms of MRKH syndrome (congenital absence of uterus and vagina). J. Negat. Results Biomed. 2006, 5, 4. [Google Scholar] [CrossRef]
  105. Acién, P.; Galán, F.; Manchón, I.; Ruiz, E.; Acién, M.; Alcaraz, A.L. Hereditary renal adysplasia, pulmonary hypoplasia and Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome: A case report. Orphanet J. Rare Dis. 2010, 5, 6. [Google Scholar] [CrossRef]
  106. Resendes, B.L.; Sohn, S.H.; Stelling, J.R.; Tineo, R.; Davis, A.J.; Gray, M.R.; Reindollar, R.H. Role for anti-Müllerian hormone in congenital absence of the uterus and vagina. Am. J. Med. Genet. 2001, 98, 129–136. [Google Scholar] [CrossRef] [PubMed]
  107. Clément-Ziza, M.; Khen-Dunlop, N.; Gonzales, J.; Crétolle-Vastel, C.; Picard, J.-Y.; Tullio-Pelet, A.; Besmond, C.; Munnich, A.; Lyonnet, S.; Nihoul-Fékété, C. Exclusion ofWNT4 as a major gene in Rokitansky-Küster-Hauser anomaly. Am. J. Med. Genet. Part A 2005, 137, 98–99. [Google Scholar] [CrossRef] [PubMed]
  108. Zenteno, J.C.; Carranza-Lira, S.; Kofman-Alfaro, S. Molecular analysis of the anti-Müllerian hormone, the anti-Müllerian hormone receptor, and galactose-1-phosphate uridyl transferase genes in patients with the May-er-Rokitansky-Küster-Hauser syndrome. Arch. Gynecol. Obstet. 2004, 269, 270–273. [Google Scholar] [CrossRef] [PubMed]
  109. Klipstein, S.; Bhagavath, B.; Topipat, C.; Sasur, L.; Reindollar, R.; Gray, M. The N314D polymorphism of the GALT gene is not associated with congenital absence of the uterus and vagina. Mol. Hum. Reprod. 2003, 9, 171–174. [Google Scholar] [CrossRef] [PubMed]
  110. Plevraki, E.; Kita, M.; Goulis, D.G.; Hatzisevastou-Loukidou, H.; Lambropoulos, A.F.; Avramides, A. Bilateral ovarian agen-esis and the presence of the testis-specific protein 1-Y-linked gene: Two new features of Mayer-Rokitansky-Küster-hauser syndrome. Fertil. Steril. 2004, 81, 689–692. [Google Scholar] [CrossRef] [PubMed]
  111. Cramer, D.W.; Goldstein, D.P.; Fraer, C.; Reichardt, J. Vaginal agenesis (Mayer-Rokitansky-Kuster-Hauser Syndrome) asso-ciated with the N314D mutation of galactose-1-phosphate uridyl transferase (GALT). Mol. Hum. Reprod. 1996, 2, 145–148. [Google Scholar] [CrossRef] [PubMed]
  112. Stévant, I.; Nef, S. Genetic Control of Gonadal Sex Determination and Development. Trends Genet. 2019, 35, 346–358. [Google Scholar] [CrossRef]
  113. Rotgers, E.; Jørgensen, A.; Yao, H.H. At the Crossroads of Fate-Somatic Cell Lineage Specification in the Fetal Gon-ad. Endocr. Rev. 2018, 39, 739–759. [Google Scholar] [CrossRef]
  114. Orvis, G.D.; Behringer, R.R. Cellular mechanisms of Müllerian duct formation in the mouse. Dev. Biol. 2007, 306, 493–504. [Google Scholar] [CrossRef]
  115. Mullen, R.D.; Behringer, R.R. Molecular genetics of Müllerian duct formation, regression and differentiation. Sex. Dev. 2014, 8, 281–296. [Google Scholar] [CrossRef]
  116. Kobayashi, A.; Shawlot, W.; Kania, A.; Behringer, R.R. Requirement of Lim1 for female reproductive tract development. Development 2004, 131, 539–549. [Google Scholar] [CrossRef] [PubMed]
  117. Zhan, Y.; Fujino, A.; MacLaughlin, D.T.; Manganaro, T.F.; Szotek, P.P.; Arango, N.A.; Teixeira, J.; Donahoe, P.K. Müllerian inhibiting substance regulates its receptor/SMAD signaling and causes mesenchymal transition of the coe-lomic epithelial cells early in Müllerian duct regression. Development 2006, 133, 2359–2369. [Google Scholar] [CrossRef] [PubMed]
  118. Arango, N.A.; Kobayashi, A.; Wang, Y.; Jamin, S.P.; Lee, H.H.; Orvis, G.D.; Behringer, R.R. A mesenchymal per-spective of Müllerian duct differentiation and regression in Amhr2-lacZ mice. Mol. Reprod. Dev. 2008, 75, 1154–1162. [Google Scholar] [CrossRef] [PubMed]
  119. Davis, R.J.; Harding, M.; Moayedi, Y.; Mardon, G. Mouse Dach1 and Dach2 are redundantly required for Müllerian duct development. Genesis 2008, 46, 205–213. [Google Scholar] [CrossRef] [PubMed]
  120. Huang, C.C.; Orvis, G.D.; Kwan, K.M.; Behringer, R.R. Lhx1 is required in Müllerian duct epithelium for uterine development. Dev. Biol. 2014, 389, 124–136. [Google Scholar] [CrossRef] [PubMed]
  121. Vainio, S.; Heikkila, M.; Kispert, A.; Chin, N.; McMahon, A.P. Female development in mammals is regulated by Wnt-4 signaling. Nature 1999, 397, 405–409. [Google Scholar] [CrossRef]
  122. Carroll, T.J.; Park, J.S.; Hayashi, S.; Majumdar, A.; McMahon, A.P. Wnt9b plays a central role in the regulation of mesen-chymal to epithelial transitions underlying organogenesis of the mammalian urogenital system. Dev.Cell 2005, 9, 283–292. [Google Scholar] [CrossRef]
  123. Arango, N.A.; Lovell-Badge, R.; Behringer, R.R. Targeted mutagenesis of the endogenous mouse Mis gene promot-er: In vivo definition of genetic pathways of vertebrate sexual development. Cell 1999, 99, 409–419. [Google Scholar] [CrossRef]
  124. Lasala, C.; Schteingart, H.F.; Arouche, N.; Bedecarrás, P.; Grinspon, R.P.; Picard, J.Y.; Josso, N.; di Clemente, N.; Rey, R.A. SOX9 and SF1 are involved in cyclic AMP-mediated upregulation of anti-Mullerian gene expression in the testicular prepubertal Sertoli cell line SMAT1. American journal of physiology. Endocrinol. Metab. 2011, 301, E539–E547. [Google Scholar] [CrossRef]
  125. Barrionuevo, F.; Bagheri-Fam, S.; Klattig, J.; Kist, R.; Taketo, M.M.; Englert, C.; Scherer, G. Homozygous Inactivation of Sox9 Causes Complete XY Sex Reversal in Mice. Biol. Reprod. 2006, 74, 195–201. [Google Scholar] [CrossRef]
  126. Kim, Y.; Bingham, N.; Sekido, R.; Parker, K.L.; Lovell-Badge, R.; Capel, B. Fibroblast growth factor receptor 2 regulates proliferation and Sertoli differentiation during male sex determination. Proc. Natl. Acad. Sci. USA 2007, 104, 16558–16563. [Google Scholar] [CrossRef] [PubMed]
  127. Bagheri-Fam, S.; Sim, H.; Bernard, P.; Jayakody, I.; Taketo, M.M.; Scherer, G.; Harley, V.R. Loss of Fgfr2 leads to par-tial XY sex reversal. Dev. Biol. 2008, 314, 71–83. [Google Scholar] [CrossRef] [PubMed]
  128. Carré, G.A.; Greenfield, A. Characterising novel pathways in testis determination using mouse genetics. Sex. Dev. 2014, 8, 199–207. [Google Scholar] [CrossRef] [PubMed]
  129. Sutton, E.; Hughes, J.; White, S.; Sekido, R.; Tan, J.; Arboleda, V.; Rogers, N.; Knower, K.; Rowley, L.; Eyre, H.; et al. Identification of SOX3 as an XX male sex reversal gene in mice and humans. J. Clin. Investig. 2011, 121, 328–341. [Google Scholar] [CrossRef] [PubMed]
  130. Ottolenghi, C.; Omari, S.; García-Ortiz, J.E.; Uda, M.; Crisponi, L.; Forabosco, A.; Pilia, G.; Schlessinger, D. Foxl2 is required for commitment to ovary differentiation. Hum. Mol. Genet. 2005, 14, 2053–2062. [Google Scholar] [CrossRef]
  131. Wilhelm, D.; Martinson, F.; Bradford, S.; Wilson, M.J.; Combes, A.N.; Beverdam, A.; Bowles, J.; Mizusaki, H.; Koopman, P. Sertoli cell differentiation is induced both cell-autonomously and through prostaglandin signaling during mammalian sex determination. Dev. Biol. 2005, 287, 111–124. [Google Scholar] [CrossRef] [PubMed]
  132. Kim, Y.; Kobayashi, A.; Sekido, R.; DiNapoli, L.; Brennan, J.; Chaboissier, M.-C.; Poulat, F.; Behringer, R.R.; Lov-ell-Badge, R.; Capel, B. Fgf9 and Wnt4 Act as Antagonistic Signals to Regulate Mammalian Sex Determination. PLoS Biol. 2006, 4, e187. [Google Scholar] [CrossRef] [PubMed]
  133. Kobayashi, A.; Stewart, C.A.; Wang, Y.; Fujioka, K.; Thomas, N.C.; Jamin, S.P.; Behringer, R.R. β-Catenin is essential for Müllerian duct regression during male sexual differentiation. Development 2011, 138, 1967–1975. [Google Scholar] [CrossRef]
  134. Deutscher, E.; Hung-Chang Yao, H. Essential roles of mesenchyme-derived beta-catenin in mouse Müllerian duct morphogenesis. Dev. Biol. 2007, 307, 227–236. [Google Scholar] [CrossRef]
  135. Ungewitter, E.K.; Yao, H.H. How to make a gonad: Cellular mechanisms governing formation of the testes and ova-ries. Sex Dev. 2013, 7, 7–20. [Google Scholar] [CrossRef]
  136. Schrader, T.J. Encyclopedia of Food Sciences and Nutrition, 2nd ed.; Academic Press: New York, NY, USA, 2003; pp. 4059–4067. [Google Scholar] [CrossRef]
  137. Liu, J.L.C. Coding or Noncoding, the Converging Concepts of RNAs. Front. Genet. 2019, 10, 496. [Google Scholar] [CrossRef]
  138. Candidate Gene. Available online: https://www.genome.gov/genetics-glossary/Candidate-Gene (accessed on 20 August 2023).
  139. Brewer, C.; Holloway, S.; Zawalnyski, P.; Schinzel, A.; FitzPatrick, D. A chromosomal deletion map of human malfor-mations. Am. J. Hum. Genet. 1998, 63, 1153–1159. [Google Scholar] [CrossRef] [PubMed]
  140. Clancy, S.; Shaw, K. DNA deletion and duplication and the associated genetic disorders. Nat. Educ. 2008, 1, 23. [Google Scholar]
  141. Cheng, Z.; Zhu, Y.; Su, D.; Wang, J.; Cheng, L.; Chen, B.; Wei, Z.; Zhou, P.; Wang, B.; Ma, X.; et al. A novel mutation of HOXA10 in a Chinese woman with a Mullerian duct anomaly. Hum. Reprod. 2011, 26, 3197–3201. [Google Scholar] [CrossRef] [PubMed]
  142. Ekici, A.B.; Strissel, P.L.; Oppelt, P.G.; Renner, S.P.; Brucker, S.; Beckmann, M.W.; Strick, R. HOXA10 and HOXA13 sequence variations in human female genital malformations including congenital absence of the uterus and vagina. Gene 2013, 518, 267–272. [Google Scholar] [CrossRef] [PubMed]
  143. Biason-Lauber, A.; De Filippo, G.; Konrad, D.; Scarano, G.; Nazzaro, A.; Schoenle, E. WNT4 deficiency—A clinical phenotype distinct from the classic Mayer–Rokitansky–Kuster–Hauser syndrome: A Case Report. Hum. Reprod. 2007, 22, 224–229. [Google Scholar] [CrossRef]
  144. Philibert, P.; Biason-Lauber, A.; Rouzier, R.; Pienkowski, C.; Paris, F.; Konrad, D.; Schoenle, E.; Sultan, C. Identification and Functional Analysis of a New WNT4 Gene Mutation among 28 Adolescent Girls with Primary Amenorrhea and Muüllerian Duct Abnormalities: A French Collaborative Study. J. Clin. Endocrinol. Metab. 2008, 93, 895–900. [Google Scholar] [CrossRef]
  145. Ravel, C.; Lorenço, D.; Dessolle, L.; Mandelbaum, J.; McElreavey, K.; Darai, E.; Siffroi, J.P. Mutational analysis of the WNT gene family in women with May-er-Rokitansky-Kuster-Hauser syndrome. Fertil Steril 2009, 91 (Suppl. S4), 1604–1607. [Google Scholar] [CrossRef]
  146. Mansouri, A.; Chowdhury, K.; Gruss, P. Follicular cells of the thyroid gland require Pax8 gene function. Nat. Genet. 1998, 19, 87–90. [Google Scholar] [CrossRef]
  147. Sauter, C.N.; McDermid, R.L.; Weinberg, A.L.; Greco, T.L.; Xu, X.; Murdoch, F.E.; Fritsch, M.K. Differentiation of murine embryonic stem cells in-duces progesterone receptor gene expression. Exp. Cell Res. 2005, 311, 251–264. [Google Scholar] [CrossRef]
  148. Lappin, T.R.; Grier, D.G.; Thompson, A.; Halliday, H.L. HOX genes: Seductive science, mysterious mechanisms. Ulster Med. J. 2006, 75, 23–31, Erratum in Ulster Med. J. 2006, 75, 135. [Google Scholar] [PubMed]
  149. Taylor, H.S. Endocrine disruptors affect developmental programming of HOX gene expression. Fertil Steril 2008, 89, e57–e58. [Google Scholar] [CrossRef] [PubMed]
  150. Aubin, J.; Lemieux, M.; Tremblay, M.; Behringer, R.R.; Jeannotte, L. Transcriptional interferences at the Hoxa4/Hoxa5 locus: Importance of correct Hoxa5 expression for the proper specification of the axial skeleton. Dev Dyn. 1998, 212, 141–156. [Google Scholar] [CrossRef]
  151. Yi, C.H.; Terrett, J.A.; Li, Q.Y.; Ellington, K.; Packham, E.A.; Armstrong-Buisseret, L.; McClure, P.; Slingsby, T.; Brook, J.D. Identification, mapping, and phylogenomic analysis of four new human members of the T box gene family: EOMES, TBX6, TBX18, and TBX19. Genomics 1999, 55, 10–20. [Google Scholar] [CrossRef] [PubMed]
  152. Coffinier, C.; Barra, J.; Babinet, C.; Yaniv, M. Expression of the vHNF1/HNF1beta homeoprotein gene during mouse organ-ogenesis. Mech. Dev. 1999, 89, 211–213. [Google Scholar] [CrossRef] [PubMed]
  153. Lokmane, L.; Heliot, C.; Garcia-Villalba, P.; Fabre, M.; Cereghini, S. vHNF1 functions in distinct regulatory circuits to con-trol ureteric bud branching and early nephrogenesis. Development 2010, 137, 347–357. [Google Scholar] [CrossRef]
  154. Anant, M.; Raj, N.; Yadav, N.; Prasad, A.; Kumar, S.; Saxena, A.K. Two Distinctively Rare Syndromes in a Case of Primary Amenorrhea: 18p Deletion and Mayer–Rokitansky–Kuster–Hauser Syndromes. J. Pediatr. Genet. 2020, 9, 193–197. [Google Scholar] [CrossRef] [PubMed]
  155. De Tomasi, L.; David, P.; Humbert, C.; Silbermann, F.; Arrondel, C.; Tores, F.; Fouquet, S.; Desgrange, A.; Niel, O.; Bole-Feysot, C.; et al. Mutations in GREB1L Cause Bilateral Kidney Agenesis in Humans and Mice. Am. J. Hum. Genet. 2017, 101, 803–814. [Google Scholar] [CrossRef]
  156. Mason, H.R.; Lake, A.C.; Wubben, J.E.; Nowak, R.A.; Castellot, J.J., Jr. The growth arrest-specific gene CCN5 is deficient in human leiomyomas and inhibits the proliferation and motility of cultured human uterine smooth muscle cells. Mol. Hum. Reprod. 2004, 10, 181–187. [Google Scholar] [CrossRef]
  157. Chen, G.; Shinka, T.; Kinoshita, K.; Yan, H.T.; Iwamoto, T.; Nakahori, Y. Roles of estrogen receptor alpha (ER alpha) in the regulation of the human Müllerian inhibitory substance (MIS) promoter. J. Med. Investig. 2003, 50, 192–198. [Google Scholar]
  158. Dell’Edera, D.; Allegretti, A.; Ventura, M.; Mercuri, L.; Mitidieri, A.; Cuscianna, G.; Epifania, A.A.; Morizio, E.; Alfonsi, M.; Guanciali-Franchi, P. Mayer-Rokitansky-Küster-Hauser syndrome with 22q11.21 microduplication: A case report. J. Med. Case Rep. 2021, 15, 208. [Google Scholar] [CrossRef] [PubMed]
  159. Wottgen, M.; Brucker, S.; Renner, S.P.; Strissel, P.L.; Strick, R.; Kellermann, A.; Wallwiener, D.; Beckmann, M.W.; Oppelt, P. Higher incidence of linked malformations in siblings of May-er-Rokitansky-Küster-Hauser-syndrome patients. Hum. Reprod. 2008, 23, 1226–1231. [Google Scholar] [CrossRef] [PubMed]
  160. Shawlot, W.; Behringer, R.R. Requirement for LIml in head-organizer function. Nature 1995, 374, 425–430. [Google Scholar] [CrossRef] [PubMed]
  161. Aydos, S.; Tükün, A.; Bökesoy, I. Gonadal dysgenesis and the Mayer-Rokitansky-Kuster-Hauser syndrome in a girl with 46,X,del(X)(pter-->q22:). Arch. Gynecol. Obstet. 2003, 267, 173–174. [Google Scholar] [CrossRef] [PubMed]
  162. Kucheria, K.; Taneja, N.; Kinra, G. Autosomal translocation of chromosomes 12q & 14q in mullerian duct failure. Indian J. Med. Res. 1988, 87, 290–292. [Google Scholar] [PubMed]
  163. Amesse, L.; Yen, F.F.; Weisskopf, B.; Hertweck, S.P. Vaginal uterine agenesis associated with amastia in a phenotypic female with a de novo 46,XX,t(8;13)(q22.1;q32.1) translocation. Clin. Genet. 1999, 55, 493–495. [Google Scholar]
  164. ACTR3B. Available online: https://www.proteinatlas.org/ENSG00000133627-ACTR3B/tissue (accessed on 15 February 2023).
  165. Leacock, S.W. Available online: https://bio.libretexts.org/@go/page/102481?pdf (accessed on 4 October 2023).
  166. Dhar, G.A.; Saha, S.; Mitra, P.; Nag Chaudhuri, R. DNA methylation and regulation of gene expression: Guardian of our health. Nucleus 2021, 64, 259–270. [Google Scholar] [CrossRef]
  167. Sandbacka, M.; Bruce, S.; Halttunen, M.; Puhakka, M.; Lahermo, P.; Hannula-Jouppi, K.; Lipsanen-Nyman, M.; Kere, J.; Ait-tomäki, K.; Laivuori, H. Methylation of H19 and its imprinted control region (H19 ICR1) in Müllerian aplasia. Fertil. Steril. 2011, 95, 2703–2706. [Google Scholar] [CrossRef]
  168. Eggermann, T.; Ledig, S.; Begemann, M.; Elbracht, M.; Kurth, I.; Wieacker, P. Search for altered imprinting marks in Mayer–Rokitansky–Küster–Hauser patients. Mol. Genet. Genom. Med. 2018, 6, 1225–1228. [Google Scholar] [CrossRef]
  169. Saso, S.; Clarke, A.; Bracewell-Milnes, T.; Saso, A.; Al-Memar, M.; Thum, M.-Y.; Yazbek, J.; Del Priore, G.; Hardiman, P.; Ghaem-Maghami, S.; et al. Psychological issues associated with absolute uterine factor infertility and attitudes of patients toward uterine transplantation. Prog Transpl. 2016, 26, 28–39. [Google Scholar] [CrossRef]
  170. Herlin, M.K.; Petersen, M.B.; Brännström, M. Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome: A comprehensive update. Orphanet J. Rare Dis. 2020, 15, 214. [Google Scholar] [CrossRef] [PubMed]
  171. Hodson, N.; Townley, L.; Earp, B.D. Removing harmful options: The law and ethics of international commercial surrogacy. Med. Law Rev. 2019, 27, 597–622. [Google Scholar] [CrossRef] [PubMed]
  172. International Federation of Fertility Societies (IFFS). IFFS surveillance 2016. Glob. Reprod. Health 2016, 1, 1–143. [Google Scholar] [CrossRef]
  173. White, P.M. Commercialization, altruism, clinical practice: Seeking explanation for similarities and differences in Califor-nian and Canadian gestational surrogacy outcomes. Womens Health Issues 2018, 28, 239–250. [Google Scholar] [CrossRef] [PubMed]
  174. Saran, J.; Padubidri, J.R. New laws ban commercial surrogacy in India. Med. Leg. J. 2020, 88, 148–150. [Google Scholar] [CrossRef]
  175. Bröannström, M.; Johannesson, L.; Bokström, H.; Kvarnströom, N.; Mölne, J.; Dahm-Kähler, P.; Enskog, A.; Milenkovic, M.; Ekberg, J.; Diaz-Garcia, C.; et al. Livebirth after uterus transplantation. Lancet 2015, 385, 607–616. [Google Scholar] [CrossRef]
  176. Jones, B.P.; Saso, S.; Bracewell-Milnes, T.; Thum, M.Y.; Nicopoullos, J.; Diaz-Garcia, C.; Friend, P.; Ghaem-Maghami, S.; Testa, G.; Johannesson, L.; et al. Human uterine transplantation: A review of outcomes from the first 45 cases. BJOG 2019, 126, 1310–1319. [Google Scholar] [CrossRef]
  177. Johannesson, L.; Enskog, A.; Mölne, J.; Diaz-Garcia, C.; Hanafy, A.; Dahm-Kähler, P.; Tekin, A.; Tryphonopoulos, P.; Morales, P.; Rivas, K.; et al. Preclinical report on allogeneic uterus transplanta-tion in non-human primates. Hum. Reprod. 2013, 28, 189–198. [Google Scholar] [CrossRef]
  178. 178 Möolne, J.; Broecker, V.; Ekberg, J.; Nilsson, O.; Dahm-Köahler, P.; Brännström, M. Monitoring of human uterus transplantation with cervical biopsies: A provisional scoring system for rejection. Am. J. Transplant. 2017, 17, 1628–1636. [Google Scholar] [CrossRef]
  179. London, N.J.; Farmery, S.M.; Will, E.J.; Davison, A.M.; Lodge, J.P. Risk of neoplasia in renal transplant patients. Lancet 1995, 346, 403–406. [Google Scholar] [CrossRef]
  180. Catsanos, R.; Rogers, W.; Lotz, M. The ethics of uterus transplantation. Bioethics 2013, 27, 65–73. [Google Scholar] [CrossRef] [PubMed]
  181. Arora, K.S.; Blake, V. Uterus transplantation: Ethical and regulatory challenges. J. Med. Ethics. 2014, 40, 396–400. [Google Scholar] [CrossRef] [PubMed]
  182. Daar, J.; Klipstein, S. Refocusing the ethical choices in womb transplantation. J. Law Biosci. 2016, 3, 383–388. [Google Scholar] [CrossRef]
Figure 1. Prisma flow diagram for study inclusions.
Figure 1. Prisma flow diagram for study inclusions.
Ijms 25 00120 g001
Table 1. Genes suspected to be involved in MRKH.
Table 1. Genes suspected to be involved in MRKH.
Genes and Location in ChromosomeReferenceMethod UsedAssociations
MEFV and IL-32-16p13.3
CMTM7-3p22.3
CCR4 3p22.3
[40]CGH and RT-qPCRIL32 and MEFV gene mutations associated with Mediterranean fever. MRKHS
BAZ2B and SLC4A10-2q24.2
KLHL18-3p21.31
PIK3CD-1p36.22
TNK2-3q29
[41]WGSMRKHS
LAMC1-1q25.3
RARA-17q21.2
HOXA10-7p15.2
PAX2-10q24–25
MMP14 and
LRP10-14q11.2
[39]WES, confirmed by Sanger sequencingMRKHS
IFTP57, HHLA2 and MYH15-3q13.13
PLA2R1-2q23-q24
ITGB6 and RBMS1-2q24.2
[42]SNP microarray analysisMRKHS
LRP10-14q11.2
FRAS1-4q21.21
CC2D2A-4p15.32
KIF14-1q32.1
RSPO4-20p13
MKKS-20p12.2
NPHP3-3q22.1
DYNC2H1-11q22.3
SPECC1L-22q11
VWF-12p13.31
[43]WESMRKHS
TBC1D1-4p14
KMT2D-12q13.12
HOXD3-2q31-37
DLG5-10q22.3
GLI3-7p14.1
HIRA-22q11.21
GATA3-10p14
LIFR-5p13.1
CLIP1-12q24.31
[44]Sanger sequencingMRKHS
PRKX-Xp22.33
HOXC8-12q13.13
[45]RT-qPCRMRKHS, Urinary malformations, skeletal malformations, and/or hearing defects.
MUC1-1q22[45,46]Array analysis, RT-qPCRMRKHS
RBM8A-1q21[21,47,48,49,50]Array CGH, MLPATAR syndrome (thrombocytopenia, absence of radius) [21,47,48,49]
WNT9B-17q21[38,39,49,51,52,53,54]Array CGH, WESMA, renal abnormalities, and cervicothoracic somite dysplasia [38]
TBX6-16p11.2[38,39,43,48,49,50,55,56,57,58,59]Array CGH. WESAutism spectrum disorders, neurological disorders, unaffected persons [49]
ACTR3B-Pseudogenes chromosomes 2, 4, 10, 16, 22 and Y[53]WESMA
LHX1-17q12[21,47,49,54,55,56,60,61,62,63,64,65,66,67,68]Array CGH, gene sequencingMA, Anomalies in the body axis formation [49], diabetes [61], learning disability [61]
TCF2/HNF1B-17q12[21,47,55,56,60,61,62,63,64,65,66,67,69,70]Array CGH, DNA sequencingMA, Renal cysts [49] diabetes [49,61], learning disability [61]
TBX1-22q11[21,47,48,55,71,72,73,74]Array CGH, FISH, MLPADiGeorge syndrome, heart defect, hypocalcemia, immunodeficiency, typical facial malformations, cognitive and behavioral disorders
WNT4-1p36.12[39,66,75,76,77]PCR sequencing, CGHHyperandrogenism (Atypical MRKHS), Gonadal dysgenesis
GREB1L-18q11.1-q11.2[45,53,78,79,80]WES, CGHMRKHS type 2 with kidney abnormalities, Twins discordant for MRKHS [45]
DOCK4-7q31.1[43,63] MRKHS
ZNF277-7q31.1[63] MRKHS
DACT1-14q23.1[81] MRKHS
DLGH1-3q29[82]Direct sequencingUnilateral agenesis, pelvic kidney
OXTR-3p25.3[83]DNA sequence analysisMRKHS
ESR1-6q25[84]DNA sequence analysisMRKHS
WT1-11p13[85]PCR
GATA4-8p23.1[85]PCR
EMX2-10q26[38,86,87]Sequence analysisMRKHS, other Müllerian fusion abnormalities
SHOX-Pseudoautosomal region Xp22. 3[39,88,89]CGHMRKHS
PBX1-1q23.3[90] MRKHS
PAX8-2q14.1[22,38,53,88]Array CGH. WESMutations have been associated with thyroid dysgenesis, thyroid follicular carcinomas, and atypical follicular thyroid adenomas [91], MRKHS
CGH = Comparative genomic hybridization; MLPA = Multiplex ligation-dependent probe amplification; WES = Whole-genome exon sequencing; WGS = Whole-genome sequencing; RT-qPCR = Reverse-transcriptase quantitative polymerase chain reaction; MA = Müllerian agenesis.
Table 2. Mechanisms of genetic changes.
Table 2. Mechanisms of genetic changes.
Chromosome NumberMechanism [Reference]Possible Gene Involved
11q31.1 Duplication (size 0.4 Mb) [55]
1q44 Deletion (size 0.32 Mb) [23,48]
1p36.12 mutations
1p36.21 deletion [23]
Mutation p.(Glu226Gly) [77]
Mutation c.1026C>T [73]
Mutation p.(Arg83Cys) [143]
Mutation c.35C>T p.(Leu12Pro) [144]
Mutation c.483C>T [145]
Mutation c.697G>A p.(Ala233Thr) [75]
Mutation g.200583493A>T [43]
1q21 Deletion (size 0.4 Mb to 4.6 Mb) [21,23,48]
1q21 Duplication (size 0.26 Mb to 0.36 Mb) [47,48]
1. KIF14 [43]
2. WNT4 is responsible for sex determination and affects the invagination of coelomic epithelial cells [121].
WNT4 mutation inhibits repression of ovarian steroid enzymes and causes abnormal expression of 17α hydroxylase and hyperandrogenism [75]
3. OR4M2, ZNF816 and PDE11A [23]
22p14 Duplication (size-0.23 Mb) [21]
2p14 Mutation c.1315G>A, p.Ala439Thr [53]
2p23.1 Duplication (size-0.21 Mb) [55]
2p24 Deletion (size-4.6 Mb) [55]
2q11.2 Duplication (size-1.3 Mb) [55]
2q24.2 Duplication [42]
2q13 Deletion (size-0.12 Mb) [21]
1. The PAX8 gene encodes a homeodomain signaling molecule, strongly expressed in the MD [146].
2. Duplication at 2q24.2 of proband MRKHS involved PLA2R1, ITGB6 and RBMS1
33p21 Duplication 0.10 Mb [60]
3p21 Mutation c.861G>A [145]
3q13 Duplication at 3q13. [42]
3q29 Deletion 0.05 Mb [60]
Mutation g.132403615G>A [43]
1. WNT7A encodes secreted signaling proteins, and is involved in the development of the anterior–posterior axis in the female reproductive tract. Coded proteins are responsible for patterning during embryogenesis. Their role in MA is uncertain.
2. May involve DLGH1, OXTR, NPHP3
44q28 Deletion 0.11 Mb [60]
4q32 Deletion 0.34 Mb [60]
4q35.2 Deletion 1.1 Mb [54]
Mutation g79204031G>A and g.15542618C>T [43]
1. FRAS1 (g79204031G>A) [43]
2. CC2D2A (g.15542618C>T) [43]
55p11 Deletion 0.40 Mb [62]
5q14.3 Deletion 0.40 Mb [62]
66p21 Duplication 0.17 Mb [60]
6q25.1 Duplication 0.42 Mb [60]
6q25.2 Duplication 0.44 Mb [60]
6q11.1 Duplication 0.41 Mb [62]
77p15.2 Hypomethylation [32]
7p14 Duplication 1.75 Mb [60]
7q31.2 Deletion 1.8 Mb [54]
Mutation g111503593C>T [43]
1. HOXA5 is a transcriptional regulator of p53 and progesterone receptor (PGR). Hypomethylation leads to overexpression, which causes overexpression of PGR [147]. Ectopic HOXA5 expression at the 5′end of the cluster might prevent normal differentiation of the MD or even regression.
2. It most likely involves Abdominal B (AbdB) homeobox genes (HOX-A9, A10, A11, and A13), required for differentiation and segmental patterning of MD [148]
3. HOXA9 is expressed in the region that becomes the oviduct [149]. Ectopic expression of HOXA5 or HOXA9 inhibits MD differentiation [150].
4. DOCK4
88p23.1 Hypomethylation [85]
8p23.1 Activating mutations [32]
GATA binding protein 4 promotes AMH production and regulates sex determination and differentiation [85]. Overproduction of AMH leads to MA.
1010q24 Duplication 0.05 Mb [60]
1111p11.12 Deletion 0.76 Mb (45)
11p 13 Hypomethylation [85]
11p 13 Activating mutation [32]
Mutation g.102985987C>T [43]
1. WT1 is a regulatory factor important for the transcription of anti-Müllerian hormone (AMH) genes. It promotes AMH expression and regulates sex determination and differentiation [85]. Activating the mutation of the gene for the AMH receptor, or the receptor, causes excessive production of AMH, leading to MRKHS [32]
2. DYNC2H1
1212q13.13 Duplication [42]
12q23 Duplication 0.16 Mb [60]
12q24 Duplication 0.12 Mb [60]
Mutation g.6085324G>A [43]
VWF
1313q21 Deletion 0.41 Mb [54]
1414q32.33 Deletion 0.46 Mb [62]
14q32.33 mutation g.23345412G>A [43]
LRP10
1515q21.1 Deletion 0.28 Mb [62]
15q26.3 Deletion 0.54 Mb [54]
Deletions at 15q11.2 [23]
1616p13.3 Increased expression [40]
16q11.2 Duplication 0.20 Mb [55]
16p11.2 Deletion (size 0.55 Mb to 0.6 Mb) [55]
Splice site mutation c.622-2A>T (g.30100162 T>A) [56]
Mutation c.484G>A(rs56098093) p.Gly162Ser [56]
Mutation c.815G>A p.Arg272Gln [56]
Mutation c.815G>A [54]
1. Genes for IL32 and MEFV.
2. TBX6 is involved in paraxial mesoderm formation and somitogenesis in human embryos [151]. Deletion induces MRKHS due to the loss of the transcription factor.
1717q12 Deletion (size 1.2 to 1.9 Mb) [21,42,47,48,55,60,62]
17q12 Missense mutation of LHX1 [21,61]
c.790C>G p.(Arg264Gly)
c. c.25dup p.(Arg9Lysfs*25)
17q21-22 mutations
c.28G>T p.Ala10Ser
c.205C>T, p.Arg69Trp [53]
c.*158C>T
17q21-22 Five Missense mutations [54]
c.472C>G p.(Gln158Glu)
c.665G>A p.(Arg222His)
c.722G>A p.(Arg24His)
c.974G>A p.(Arg325His)
c.1029C>A p.(Cys343*)
1. Involves the LHX1(LIM homeobox protein 1) gene, which is a transcription factor necessary for the formation of the Müllerian duct-derived uterine and vaginal epithelia [116].
2. HNF1B is [Pit–Oct–Unc homeodomain-containing transcription factor that is frequently expressed in the Müllerian duct during development [152]. It positively regulates the expression of LHX1, PAX2, and WNT9B [153].
3. Involves mutations in WNT9B, which acts upstream of another Wnt4. It is responsible for the caudal extension of the Müllerian duct and the organization of the urogenital system [122].
1818q23 Duplication 0.20 Mb [62]
18p Deletion [154]
18q11.1-q11.2 mutation c.4665T>A, p.Tyr1555
GREB1L is a target gene in the retinoic acid signaling pathway, which is highly expressed in the developing fetal human kidney and involved in the early metanephros and genital development [155].
1919q13.31 deletion [23]OR2T2 [23]
2020q13.12 Deletion
Mutations g.941074G>A and g.10393439C>A [43]
1. WISP2 is significant in smooth muscle cell proliferation and migration, and is induced by estrogen in the uterus [156]. Estrogen regulates AMH expression levels [157], and overexposure to estrogen during development activates AMH promotors [32].
2. RSPO4
3. MKKS
2222q11 Deletion (size 0.39 Mb to 2.6 Mb) [55,61,71,73]
Duplication (0.6 Mb–3.5 Mb) [47,61,158]
Mutations g.24720297G>A and g.24718408G>A [43]
1. TBX1
2. SPECC1L
XXp11.1 Deletion 0.12 Mb [60]
Xp11.3 Duplication 0.24 Mb [60]
Xp22 Duplication (0.07 to 0.36) [21,45,47,48,159]
Xq21.31 Deletion 1 Mb [160]
Xq deletion [161]
Xq22.3 Duplication 0.09 Mb [60]
Xq22.3 Microdeletion at Xp22.33 [42]
1. May involve the PRKX gene, encoding for a serine/threonine kinase implicated in renal epithelium morphogenesis [45].
2. May involve the SHOX gene, which encodes a transcription factor responsible for skeletal growth. The exact mechanism is unknown.
8,13t(8;13) (q12;q14) translocationTranslocation causes MRKHSS with or without renal hypoplasia [162].
t(8;13)(q22.1;q32.1) translocationTranslocation causes MRKHS with limb, breast, and urinary functional defects [163]
3,16t(3;16) (p22.3;p13.3) translocationTranslocation causes MRKHS [40,98]
7,14t(7;14)(q32;q32) translocationTranslocation seen in MRKHS [98]
2, 4, 10, 16, 22 and Yc.1066G>A, p.Gly356Arg [53]ACTR3B encodes a member of the actin-related proteins and plays a role in the organization of the actin cytoskeleton [164]. ACTR3B can have pseudogenes in more than one chromosome.
A glossary of the genes can be found in the supplementary materials; MD = Müllerian duct.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dube, R.; Kar, S.S.; Jhancy, M.; George, B.T. Molecular Basis of Müllerian Agenesis Causing Congenital Uterine Factor Infertility—A Systematic Review. Int. J. Mol. Sci. 2024, 25, 120. https://doi.org/10.3390/ijms25010120

AMA Style

Dube R, Kar SS, Jhancy M, George BT. Molecular Basis of Müllerian Agenesis Causing Congenital Uterine Factor Infertility—A Systematic Review. International Journal of Molecular Sciences. 2024; 25(1):120. https://doi.org/10.3390/ijms25010120

Chicago/Turabian Style

Dube, Rajani, Subhranshu Sekhar Kar, Malay Jhancy, and Biji Thomas George. 2024. "Molecular Basis of Müllerian Agenesis Causing Congenital Uterine Factor Infertility—A Systematic Review" International Journal of Molecular Sciences 25, no. 1: 120. https://doi.org/10.3390/ijms25010120

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop