ArticlePDF Available

FMRP targets distinct mRNA sequence elements to regulate protein expression

Authors:

Abstract and Figures

Fragile X syndrome (FXS) is a multi-organ disease that leads to mental retardation, macro-orchidism in males and premature ovarian insufficiency in female carriers. FXS is also a prominent monogenic disease associated with autism spectrum disorders (ASDs). FXS is typically caused by the loss of fragile X mental retardation 1 (FMR1) expression, which codes for the RNA-binding protein FMRP. Here we report the discovery of distinct RNA-recognition elements that correspond to the two independent RNA-binding domains of FMRP, in addition to the binding sites within the messenger RNA targets for wild-type and I304N mutant FMRP isoforms and the FMRP paralogues FXR1P and FXR2P (also known as FXR1 and FXR2). RNA-recognition-element frequency, ratio and distribution determine target mRNA association with FMRP. Among highly enriched targets, we identify many genes involved in ASD and show that FMRP affects their protein levels in human cell culture, mouse ovaries and human brain. Notably, we discovered that these targets are also dysregulated in Fmr1(-/-) mouse ovaries showing signs of premature follicular overdevelopment. These results indicate that FMRP targets share signalling pathways across different cellular contexts. As the importance of signalling pathways in both FXS and ASD is becoming increasingly apparent, our results provide a ranked list of genes as basis for the pursuit of new therapeutic targets for these neurological disorders.
Content may be subject to copyright.
FMR1 targets distinct mRNA sequence elements to regulate
protein expression
Manuel Ascano Jr.1, Neelanjan Mukherjee2,5, Pradeep Bandaru1, Jason B. Miller1, Jeff
Nusbaum, David L. Corcoran2, Christine Langlois3, Mathias Munschauer, Scott Dewell4,
Markus Hafner1, Zev Williams1,3, Uwe Ohler2,5,*, and Thomas Tuschl1,*
1Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller
University, New York, NY 10065, USA
2Institute for Genome Sciences and Policy, Duke University, Durham, NC 27708, USA
3Program for Early and Recurrent Pregnancy Loss, Department of Obstetrics & Gynecology and
Women's Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
4Genomics Resource Center, The Rockefeller University, New York, NY, 10065, USA
Abstract
Fragile-X Syndrome (FXS) is a multi-organ disease leading to mental retardation, macro-
orchidism in males, and premature ovarian insufficiency in female carriers. FXS is also a
prominent monogenic disease associated with autism spectrum disorders (ASD). FXS is typically
caused by the loss of FRAGILE X-MENTAL RETARDATION 1 (FMR1) expression, which
encodes for the RNA-binding protein (RBP), FMRP. We report the discovery of distinct RNA
recognition elements (RREs) that correspond to the two independent RNA binding domains of
FMRP, and the binding sites within the mRNA targets for wild-type and I304N mutant FMRP
isoforms and its paralogs, FXR1 and FXR2. RRE frequency, ratio, and distribution determine
target mRNA association with FMRP. Among highly-enriched targets, we identified many genes
involved in ASD and demonstrate that FMRP affects their protein levels in cell culture, mice, and
human brain. Unexpectedly, we discovered that these targets are also dysregulated in Fmr1-/-
mouse ovaries, showing signs of premature follicular overdevelopment. These results indicate that
FMRP targets shared signaling pathways across different cellular contexts. As it is become
increasingly appreciated that signaling pathways are important to FXS and ASD, our results here
Users may view, print, copy, download and text and data- mine the content in such documents, for the purposes of academic research,
subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms
*Corresponding authors Thomas Tuschl ttuschl@rockefeller.edu.
5Present address: The Berlin Institute for Medical Systems Biology, Max Delbrück Center, Berlin-Buch, Germany
CONTRIBUTIONS
M.A designed, executed, supervised, and interpreted experiments. N.M., P.B., and D.L.C. carried out the sequence alignment,
annotation, and PARalyzer pipeline. N.M. and P.B. performed the computational analysis on the RIP-chip. M.A., J.B.M., and J.N.
purified FMRP proteins, performed the EMSAs, and carried out the quantitative Westerns and analyses. M.A. and M.M. performed
the RIP-chips. S.D. assisted in the Illumina sequencing of all PAR-CLIP libraries. M.H. helped in the initial PAR-CLIP experiments.
C.L. and Z.W. carried out and analyzed mouse experiments. U.O. supervised computational efforts. T.T. supervised and helped in the
design of experiments. M.A. and T.T. wrote the manuscript.
COMPETING FINANCIAL INTEREST
T.T. is co-founder and scientific advisor to Alnylam Pharmaceuticals and Regulus Therapeutics.
HHS Public Access
Author manuscript
Nature. Author manuscript; available in PMC 2013 June 20.
Published in final edited form as:
Nature. 2012 December 20; 492(7429): 382–386. doi:10.1038/nature11737.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
provide a molecular guide towards the pursuit of novel therapeutic targets for these neurological
disorders.
INTRODUCTION
Most clinical cases of FXS are a result of a hyper-expansion and methylation of CGG
repeats within the promoter of FMR1, leading to a loss of its expression1-3. The FMR1 RBP
family has three members, FMR1, FXR1, and FXR2, which possess two centrally located
KH domains and a C-terminal arginineglycine (RG)-rich region implicated in mRNA
binding 4-7. FMR1 encodes for multiple protein isoforms, but is predominantly expressed as
a 69 kDa protein (isoform 7)8,9. Isoform (iso) 1 and six other alternative splice variants
include exon 12, with iso1 encoding the full-length protein (71 kDa). Exon 12 insertion
lengthens the second KH (KH2) RBD, possibly influencing FMR1 RNA-binding specificity
or affinity. The I304N mutation, described in a FXS patient, is also located in the KH2 and
is reported to attenuate association with RNA and polysomes10-12. FMR1 proteins are
implicated in various RNA processes including RNA subcellular localization by facilitating
nucleo-cytoplasmic shuttling13 and association with motor proteins14-16. FMR1 proteins
were also suggested to mediate translational regulation12,17. Given the critical role of FMR1
in human cognition and premature ovarian insufficiency18,19, intensive efforts towards the
identification of its RNA targets have been employed, with the goal that their discovery
would shed light upon the array of related disorders and provide options for molecular
therapy18-26. No precise RRE has been defined and very few bona fide mRNA targets are
confirmed27.
RESULTS AND DISCUSSION
To identify the binding sites of FMR1 family proteins (Fig. 1a and Supplementary Fig. 1),
we first compared photocrosslinking methods28-30 using stable FLAG-HA FMR1 iso7
expressing HEK293 cells (Fig. 1b), as these cells and human brain share 90% of expressed
genes based on a comparison of existing RNAseq datasets31-33 (Supplementary Fig. 2). The
difference in FMR1 levels between the experimental system and brain is 1.3 fold, as
calculated using RNAseq data and the quantitated expression of FMR1 in our stable cells.
We found that 4SU PAR-CLIP provided the highest yield of crosslinked RNAs, and used
this approach for all FMR1 family proteins (Fig. 1c). cDNA libraries were generated after
PAR-CLIP and Illumina-sequenced (Supplementary Table 1). Genome-aligned reads were
grouped by PARalyzer34 to identify segments of RNA that represented peaks of T-to-C
conversion, termed binding sites. PARalyzer separated closely spaced binding sites
connected by overlapping reads and yielded a median RNA segment length of 33 nt
(Supplementary Fig. 3). FMRP iso1 and 7 bound to approx. 80,000-100,000 sites, of which
> 85% mapped to ~6,000 mRNAs (Supplementary Tables 1, 2, and https://
fmrp.rockefeller.edu). FXR1 and FXR2 protein binding sites are comprised within FMRP
binding sites with an overlap of 95% (Supplementary Table 3).
Nearly all mRNA binding sites were located in exons (>90%) (Fig. 2a) and distributed
between CDS and 3'UTR (>95%, total) with slightly more CDS sites, similar to distributions
seen for other cytoplasmic RBPs28. The computational sequence analysis method cERMIT35
Ascano et al. Page 2
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
revealed two major RREs, ACUK and WGGA (K=G/U, and W=A/U) (Fig 2b,
Supplementary Fig. 4). Together, ACUK and WGGA RREs were found in ≥50% of mRNA
binding sites in iso1 and iso7; they occurred exclusively or together within the same binding
site (Fig. 2c). Remaining binding sites typically contained close derivatives of either RRE.
We performed electrophoretic mobility shift assays (EMSAs) to test RNAs representing
FMRP binding sites using recombinant FMRP purified from Sf9 cells (Supplementary Fig. 5
and 6). FMRP target sites were selected based on whether they contained ACUK, WGGA or
Mixed (ACUK/WGGA) RREs (Supplementary Table 4). Testing RNAs of various lengths,
we found that oligonucleotide lengths of ≥45 nt were required to observe gel shifts and reach
dissociation constants below 0.5 μM, suggesting RNA backbone contacts outside the RREs
contribute to the association in vitro. WGGA-containing RNAs exhibited the widest range
and strongest affinities, generally correlating with the number of RREs within a PARalyzer-
defined binding site. An RNA segment containing nine WGGAs bound almost 2 orders of
magnitude tighter than those containing one WGGA, whereas binding of ACUK-containing
RNAs varied only 5-fold. EMSAs using RNAs representing target sites within PPP2CA, and
UBE3A are shown (Fig. 3a).
We also tested sites in APP and FMR1 mRNA (Supplementary Fig. 7), two previously
identified mRNA targets. APP was originally discovered as an FMRP target based on a
predicted G-quartet region7 although the actual site was subsequently identified in vitro at an
upstream segment36, which was identified here (APP Site 1) as containing WGGA. FMRP
targets its own mRNA though it was an association only observed in vitro5,6,37 and in
immunoprecipitates38.
Recombinant I304N iso1 FMRP showed a ~10-fold average decrease in its affinity towards
ACUK-containing RNAs compared to wt FMRP iso1 (Fig. 3b and Supplementary Table 5).
We characterized binding to wt and mutant RNA sequences derived from an NF1 mixed
RRE site (Fig. 3c). I304N FMRP bound wt and NF1 ACUK(-) RNAs similarly, whereas wt
FMRP showed a two-fold reduction in affinity for NF1 ACUK(-) RNA. Additionally, wt
FMRP bound NF1 ACUK, WGGA(-) RNA similar to I304N FMRP for NF1 WGGA(-)
RNA. Our results indicate that the KH2 domain associates with the ACUK RRE. Since
FMRP associates with mRNAs at more than one binding site, its association at multiple sites
will impact the final regulatory effect. We compared the distribution of RREs in I304N
FMRP vs. wt FMRP PAR-CLIPs and found a transcriptome-wide depletion in the recovery
of sequence reads for ACUK binding sites for both I304N isoforms, consistent with the
biochemistry (Fig. 3d). Interestingly, the RRE fractional distribution of I304N FMRP iso1
was similar to wt FMRP iso7, suggesting that alteration of the KH2 domain by mutation or
exon insertion affects binding-site selectivity. Taken together, the biochemical and PAR-
CLIP results with I304N FMRP indicate that we identified the natural target sites of the
FMRP KH2 domain.
To rank FMRP targets, we measured their enrichment in RIP-chip, which would indicate
stable interactions. 3,593 PAR-CLIP-identified targets showed enrichment by RIP-chip, of
which 939 genes were two- to six-fold enriched; 646 transcripts were two-fold enriched but
not identified as PAR-CLIP targets. We used binding-site information obtained by PAR-
Ascano et al. Page 3
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
CLIP to infer the salient features for stable association in RIP-Chip (Fig. 4, Supplementary
Fig. 8, and Supplementary Table 6). Increasing frequency of WGGA- and ACUK-
containing elements led to greater RIP-chip enrichment, in agreement with in vitro affinity
measurements. On average, top targets contained more RRE binding sites (18 per transcript)
compared to the least-enriched targets (13 per transcript). Top-ranked targets had a CDS:
3'UTR binding site ratio of 3.7:1 compared to bottom-ranked targets that had 1:2.
Transcripts with ACUK:WGGA RRE ratios <1 were the most significantly enriched
population. Importantly, we identified 93 genes independently implicated in ASD among the
highly-enriched FMRP targets (Fig. 4d and Supplementary Table 7), which is striking given
the relationship of FXS with ASD20,21,39.
Enrichment of RNAs in RIP-chip depends on the saturation of target sites with FMRP. To
estimate the yield of saturation, protein copy number, target mRNA copies, and the number
of binding sites within those transcripts have to be accounted. We determined endogenous
and co-expressed FLAG-HA-tagged FMR1 protein copy numbers to be approx. 60,000 and
70,000 molecules per cell, respectively, using quantitative Western blotting and reference
recombinant protein. Each cell contains 20 pg total RNA, of which 4% are the approx. 1
Mio mRNA molecules/cell. Considering their relative abundance based on HEK293
RNAseq, we estimate that an equal distribution of FMRP would occupy 6% of binding sites/
cell among its 6,000 target transcripts, or up to 20% if FXR1 and 2 protein estimates are
included. Since PAR-CLIP-identified targets had varying enrichment, the association of
FMRP with them is not uniform. The ~3,500 transcripts enriched in RIP-chip are estimated
to have at least 18% FMRP binding site occupancy, whereas top-ranked 900 genes (two-fold
enriched) potentially exhibit 78%. The presence of multiple binding sites within targets
suggests that multiple FMR1 family proteins bind each transcript to influence their
regulation.
To assess the impact of FMR1 binding sites on mRNA stability, siRNA knockdown of
FMR1 or the FMR1 family was performed and mRNA expression profiles were analysed by
microarray. We found no evidence for FMR1 affecting target mRNA abundance (data not
shown).
A panel of FMR1-targets were selected based on enrichment in RIP-Chip, low-to-
intermediate expression in RNAseq, similar abundance in human brain (using published
microarray31 and RNAseq datasets32,33) and with documented neurological and human
disease relevance, then analysed them by quantitative Western blot to determine their
protein levels as a function of FMRP expression (Fig. 4e). FXR2, HUWE1, KDM5C, and
MTOR protein levels, among others, showed up to 30% reduction in protein levels upon
expression of FMR1, in HEK293. We analysed lysates prepared from human postmortem
brains. Four FXS brains (Supplementary Fig. 9) were available with age/sex/anatomic-
matched controls from pre-frontal cortical, hippocampus, and cerebellar regions. While only
four of eight antibodies yielded quantifiable bands in brain lysates, we observed a general
trend of elevated target protein expression levels in FXS brains. This is the inverse of FMR1
overexpression effects in HEK293, and consistent with FMRP affecting the protein levels of
its mRNA targets.
Ascano et al. Page 4
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
The mRNA targets identified here are from a human transcriptome where the vast majority
of genes are comparably expressed in human brain (Supplementary Fig. 2). We discovered
ASD-related and numerous other genes implicated in neuronal disorders associated with
FXS and validated representatives by EMSA, RIP-chip, and immunoblot. We found genes
involved in Angelman (AS), Prader-Willi, Rett, and Cornelia de Lange Syndromes.
Interestingly, the ASD and AS-associated gene UBE3A ubiquitinates ARC and SACS140;
ARC is a well-known target and here we identify SACS1 as a targeted transcript. These
findings potentially provide the molecular link to tie together elements of clinically
overlapping disorders, principally setting a molecular target framework for characterizing
the connections between FXS and its associated phenotypes.
Although FX-related diseases are primarily considered CNS disorders, at least two other
target organs are affected, the testes and ovaries. We reasoned that changes in FMR1
expression lead to dysregulation of largely overlapping sets of targets shared across all
affected organs. Thus dysregulated genes and pathways in brain might also contribute to
phenotypes in testes and ovary. We therefore examined the ovaries of Fmr1-/- mice41 since
CNS and macroorchidism phenotypes were reported, yet ovary development had largely
been under-investigated. Ovaries from Fmr1-/- mice were markedly larger by 3 weeks post-
birth compared to wt controls (Fig. 5a-b). At 12 and 18 wks post-birth, KO ovaries were
22% and 72% larger by mass compared to age-matched controls, respectively. Importantly,
we found increased protein levels of Tsc2, Sash1 and Mtor (Fig. 5c). As it is independently
known that the Mtor pathway can regulate ovarian development, it is tempting to conclude
that increased activity, in the absence Fmr1 expression, contributes to enlarged ovaries
histologically consistent with precocious follicular development. These observations suggest
that Fmr1-KO mice have the potential to model FXS-related premature ovarian
insufficiency in which it remains unclear whether elevated FMR1 mRNA or the observed
reduction of FMRP protein itself is causative in female carriers affected by this disease.
Elevated signal transduction activity of the Mtor pathway42,43 was reported in Fmr1 KO-
mice, and was attributed to Fmrp targeting of Pik3ca mRNA. Indeed, PAR-CLIP identified
several FMRP binding sites within the PIK3CA transcript. However, we find that it is a less-
enriched RIP-chip target compared to MTOR and TSC2, whose protein levels appear
regulated in an FMRP-dependent manner. Interestingly, recent evidence demonstrated that
Tsc mutant mice44-46, which have increased mTOR activity, had impaired mGluR-LTD and
protein synthesis compared to Fmr1-/- mice; crossing Tsc2+/- with Fmr1-/y mice corrected
the phenotypes44. Given our results we suspect that in Tsc2+/-, Fmr1-/y mice, Tsc2 and Mtor
protein levels (among others) were elevated, correcting the balance of protein expression
and leading to the reversal of phenotypes observed. The reported model44 by which separate
pools of mRNA are differentially regulated by partially convergent pathways in FMRP
(in)dependent ways, remains unclear. This is in part because FMRP associates with
transcripts of ERK pathway components as well. Therapeutic targeting of the MTOR
pathway has become an important goal – but must be further guided by additional functional
analysis, particularly of FMRP targets upstream and downstream of MTOR and
interconnected signaling pathways (Supplementary Fig. 10). Combined, our validation work
in Fmr1-KO mouse ovaries and in human brain demonstrate that the effect of FMRP
Ascano et al. Page 5
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
binding to specific target genes identified in cell culture is extensible to physiologically
relevant contexts.
METHODS SUMMARY
Methods are described in greater detail within Supplementary Information. FVB129P2
Fragile-X mice were a kind gift from Dr. Suzanne Zukin (Albert Einstein College of
Medicine). Gateway plasmids (Invitrogen) generated in this study will be deposited in
addgene.org. FlpIn T-Rex HEK293 (Invitrogen) inducible-stable cell lines were generated
per manufacturer's instrutions. The titers, source and use of antibodies used in this study are
listed in Supplementary Information. PAR-CLIP was performed essentially as described,
except that the second RNase T1 digestion was omitted following the IP. Recombinant wt
and mutant FMRP iso1 proteins were purified using baculoviral expression system
(Invitrogen). Electrophoretic mobility shift assays and Western blots were quantified using
ImageGauge (Fuji). Parameters of computation analyses are described in Supplementary
Information and within the relevant sections within https://fmrp.rockefeller.edu/. Relevant
datasets, including raw data are available at https://fmrp.rockefeller.edu/ and GEO
(GSE39686).
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
ACKNOWLEDGEMENTS
Human tissue was obtained from the NICHD Brain and Tissue Bank for Developmental Disorders at the University
of Maryland, Baltimore, MD. We would like to thank the following members of the Tuschl lab for their support and
assistance: Greg Wardle, Dr. Neil Renwick, and Dr. Iddo Ben-Dov. We would like to thank Dr. Jack Keene for his
invaluable advice throughout the project. We would like to acknowledge Dr. Mohsen Khorshid, Dr. Lukas Burger,
and Dr. Mihaela Zavolan for analyzing PAR-CLIP data at the initial stages of the project and discussions. We
would like to thank the MSKCC in-situ core for their assistance with the mouse histology. Finally, we would like to
express our appreciation for all members of the Tuschl laboratory for their assistance and collegiality. This work
was supported, in part, by the following agencies: NIH/NCRR/RU CCTS (M.A., UL1RR024143), Simons
Foundation Autism Research Initiative (T.T., CEN5300891) and NIH (T.T., R01 MH080442).
REFERENCES CITED
1. Verkerk AJ, et al. Identification of a gene (FMR-1) containing a CGG repeat coincident with a
breakpoint cluster region exhibiting length variation in fragile X syndrome. Cell. 1991; 65:905–914.
[PubMed: 1710175]
2. Kremer EJ, et al. Mapping of DNA instability at the fragile X to a trinucleotide repeat sequence
p(CCG)n. Science (New York, N.Y. 1991; 252:1711–1714.
3. Vincent A, et al. Abnormal pattern detected in fragile-X patients by pulsed-field gel electrophoresis.
Nature. 1991; 349:624–626. [PubMed: 1672039]
4. Siomi H, Siomi MC, Nussbaum RL, Dreyfuss G. The protein product of the fragile X gene, FMR1,
has characteristics of an RNA-binding protein. Cell. 1993; 74:291–298. [PubMed: 7688265]
5. Ashley CTJ, Wilkinson KD, Reines D, Warren ST. FMR1 protein: conserved RNP family domains
and selective RNA binding. Science (New York, N.Y. 1993; 262:563–566.
6. Brown V, et al. Purified recombinant Fmrp exhibits selective RNA binding as an intrinsic property
of the fragile X mental retardation protein. J. Biol. Chem. 1998; 273:15521–15527. [PubMed:
9624140]
Ascano et al. Page 6
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
7. Darnell JC, et al. Fragile X mental retardation protein targets G quartet mRNAs important for
neuronal function. Cell. 2001; 107:489–499. [PubMed: 11719189]
8. Ashley CT, et al. Human and murine FMR-1: alternative splicing and translational initiation
downstream of the CGG-repeat. Nat. Genet. 1993; 4:244–251. [PubMed: 8358432]
9. Verkerk AJ, et al. Alternative splicing in the fragile X gene FMR1. Hum. Mol. Genet. 1993; 2:399–
404. [PubMed: 8504300]
10. De Boulle K, et al. A point mutation in the FMR-1 gene associated with fragile X mental
retardation. Nat. Genet. 1993; 3:31–35. [PubMed: 8490650]
11. Siomi H, Choi M, Siomi MC, Nussbaum RL, Dreyfuss G. Essential role for KH domains in RNA
binding: impaired RNA binding by a mutation in the KH domain of FMR1 that causes fragile X
syndrome. Cell. 1994; 77:33–39. [PubMed: 8156595]
12. Feng Y, et al. FMRP associates with polyribosomes as an mRNP, and the I304N mutation of
severe fragile X syndrome abolishes this association. Mol. Cell. 1997; 1:109–118. [PubMed:
9659908]
13. Fridell RA, Benson RE, Hua J, Bogerd HP, Cullen BR. A nuclear role for the Fragile X mental
retardation protein. EMBO J. 1996; 15:5408–5414. [PubMed: 8895584]
14. Ling SC, Fahrner PS, Greenough WT, Gelfand VI. Transport of Drosophila fragile X mental
retardation protein-containing ribonucleoprotein granules by kinesin-1 and cytoplasmic dynein.
Proc. Nat. Acad. Sci. 2004; 101:17428–17433. [PubMed: 15583137]
15. Davidovic L, et al. The fragile X mental retardation protein is a molecular adaptor between the
neurospecific KIF3C kinesin and dendritic RNA granules. Hum. Mol. Genet. 2007; 16:3047–3058.
[PubMed: 17881655]
16. Dictenberg JB, Swanger SA, Antar LN, Singer RH, Bassell GJ. A Direct Role for FMRP in
Activity-Dependent Dendritic mRNA Transport Links Filopodial-Spine Morphogenesis to Fragile
X Syndrome. Dev. Cell. 2008; 14:926–939. [PubMed: 18539120]
17. Corbin F, et al. The fragile X mental retardation protein is associated with poly(A)+ mRNA in
actively translating polyribosomes. Hum. Mol. Genet. 1997; 6:1465–1472. [PubMed: 9285783]
18. Oostra BA, Willemsen R. FMR1: a gene with three faces. Biochim. Biophys. Acta. 2009;
1790:467–477. [PubMed: 19233246]
19. De Rubeis S, Fernández E, Buzzi A, Di Marino D, Bagni C. Molecular and cellular aspects of
mental retardation in the Fragile X syndrome: from gene mutation/s to spine dysmorphogenesis.
Adv. Exp. Med. Biol. 2012; 970:517–551. [PubMed: 22351071]
20. Gross C, Berry-Kravis EM, Bassell GJ. Therapeutic strategies in fragile X syndrome: dysregulated
mGluR signaling and beyond. Neuropsychopharmacology. 2012; 37:178–195. [PubMed:
21796106]
21. Hagerman R, Lauterborn J, Au J, Berry-Kravis E. Fragile X syndrome and targeted treatment
trials. Results Probl Cell Differ. 2012; 54:297–335. [PubMed: 22009360]
22. Darnell JC, et al. FMRP Stalls Ribosomal Translocation on mRNAs Linked to Synaptic Function
and Autism. Cell. 2011; 146:247–261. [PubMed: 21784246]
23. Krueger DD, Bear MF. Toward fulfilling the promise of molecular medicine in fragile X
syndrome. Annu. Rev. Med. 2011; 62:411–429. [PubMed: 21090964]
24. Pfeiffer BE, Huber KM. The state of synapses in fragile X syndrome. Neuroscientist. 2009;
15:549–567. [PubMed: 19325170]
25. Belmonte MK, Bourgeron T. Fragile X syndrome and autism at the intersection of genetic and
neural networks. Nature neuroscience. 2006; 9:1221–1225. [PubMed: 17001341]
26. Brown V, et al. Microarray identification of FMRP-associated brain mRNAs and altered mRNA
translational profiles in fragile X syndrome. Cell. 2001; 107:477–487. [PubMed: 11719188]
27. Bassell GJ, Warren ST. Fragile X syndrome: loss of local mRNA regulation alters synaptic
development and function. Neuron. 2008; 60:201–214. [PubMed: 18957214]
28. Hafner M, et al. Transcriptome-wide Identification of RNA-Binding Protein and MicroRNA
Target Sites by PAR-CLIP. Cell. 2010; 141:129–141. [PubMed: 20371350]
29. Licatalosi DD, et al. HITS-CLIP yields genome-wide insights into brain alternative RNA
processing. Nature. 2008; 456:464–469. [PubMed: 18978773]
Ascano et al. Page 7
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
30. Ascano M, Hafner M, Cekan P, Gerstberger S, Tuschl T. Identification of RNA-protein interaction
networks using PAR-CLIP. WIREs RNA. 2012; 3:159–177. [PubMed: 22213601]
31. Su AI, et al. A gene atlas of the mouse and human protein-encoding transcriptomes. Proc. Nat.
Acad. Sci. 2004; 101:6062–6067. [PubMed: 15075390]
32. Wang ET, et al. Alternative isoform regulation in human tissue transcriptomes. Nature. 2008;
456:470–476. [PubMed: 18978772]
33. Kishore S, et al. A quantitative analysis of CLIP methods for identifying binding sites of RNA-
binding proteins. Nat. Meth. 2011; 8:559–564.
34. Corcoran DL, et al. PARalyzer: definition of RNA binding sites from PAR-CLIP short-read
sequence data. Genome Biol. 2011; 12:R79. [PubMed: 21851591]
35. Georgiev S, et al. Evidence-ranked motif identification. Genome Biol. 2010; 11:R19. [PubMed:
20156354]
36. Westmark CJ, Malter JS. FMRP mediates mGluR5-dependent translation of amyloid precursor
protein. PLoS Biol. 2007; 5:e52. [PubMed: 17298186]
37. Schaeffer C, et al. The fragile X mental retardation protein binds specifically to its mRNA via a
purine quartet motif. EMBO J. 2001; 20:4803–4813. [PubMed: 11532944]
38. Ceman S, Brown V, Warren ST. Isolation of an FMRP-associated messenger ribonucleoprotein
particle and identification of nucleolin and the fragile X-related proteins as components of the
complex. Mol. Cell. Biol. 1999; 19:7925–7932. [PubMed: 10567518]
39. Bhakar AL, Dölen G, Bear MF. The Pathophysiology of Fragile X (and What It Teaches Us about
Synapses). Annu Rev Neurosci. 2012 doi:10.1146/annurev-neuro-060909-153138.
40. Greer PL, et al. The Angelman Syndrome protein Ube3A regulates synapse development by
ubiquitinating arc. Cell. 2010; 140:704–716. [PubMed: 20211139]
41. Fmr1 knockout mice: a model to study fragile X mental retardation. The Dutch-Belgian Fragile X
Consortium. Cell. 1994; 78:23–33. [PubMed: 8033209]
42. Gross C, et al. Excess phosphoinositide 3-kinase subunit synthesis and activity as a novel
therapeutic target in fragile X syndrome. J Neurosci. 2010; 30:10624–10638. [PubMed: 20702695]
43. Sharma A, et al. Dysregulation of mTOR signaling in fragile X syndrome. J Neurosci. 2010;
30:694–702. [PubMed: 20071534]
44. Auerbach BD, Osterweil EK, Bear MF. Mutations causing syndromic autism define an axis of
synaptic pathophysiology. Nature. 2011; 480:63–68. [PubMed: 22113615]
45. Bateup HS, Takasaki KT, Saulnier JL, Denefrio CL, Sabatini BL. Loss of Tsc1 in vivo impairs
hippocampal mGluR-LTD and increases excitatory synaptic function. J Neurosci. 2011; 31:8862–
8869. [PubMed: 21677170]
46. Chévere-Torres I, et al. Metabotropic glutamate receptor-dependent long-term depression is
impaired due to elevated ERK signaling in the ΔRG mouse model of tuberous sclerosis complex.
Neurobiol. Dis. 2012; 45:1101–1110. [PubMed: 22198573]
47. Spitzer J, et al. PAR-CLIP (Photoactivatable Ribonucleoside-Enhanced Crosslinking and
Immunoprecipitation) - a step-by-step protocol to the transcriptome-wide identification of binding
sites of RNA-binding proteins. Meth. Enzymol. 2010
48. Landthaler M, et al. Molecular characterization of human Argonaute-containing ribonucleoprotein
complexes and their bound target mRNAs. RNA (New York, N.Y. 2008; 14:2580–2596.
49. Gautier L, Cope L, Bolstad BM, Irizarry RA. affy--analysis of Affymetrix GeneChip data at the
probe level. Bioinformatics. 2004; 20:307–315. [PubMed: 14960456]
Ascano et al. Page 8
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Figure 1. PAR-CLIP of FMR1 family proteins
a, FMR1 family proteins comprise two type-I KH domains (cyan). FMRP isoform 1 and 7
(iso1 and 7) vary by the presence of exon 12 (black) within KH2. The I304N mutation is
located within the KH2 domain (red asterisk). The RG-rich region (orange bars) is also
implicated in RNA-binding. The lengths of proteins in amino acids are indicated. We
established stable inducible cell lines expressing FLAG-HA epitope-tagged wt and I304N
mutants of FMR1 (iso1 and 7), and its homologs FXR1 and 247. b, RNA-FMRP
crosslinking comparing CLIP (254 nm) to 4SU-or 6SG-PAR-CLIP (365 nm). RNA-
radiolabeled FLAG immunoprecipitates (IPs) of lysates from crosslinked FLAG-HA-
FMRP-iso7-expressing HEK293 cells were separated by SDS-PAGE. The migrations of
protein mass standards are indicated. Enrichment of radiolabelled RNA covalently bound to
FLAG-HA-FMRP (arrow) was determined after normalizing by Western blot analysis (not
shown). c, 4SU-PAR-CLIP of FMR1 family proteins analogous to (b).
Ascano et al. Page 9
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Figure 2. Analysis of FMR1 family protein mRNA binding sites
a, Distribution of binding sites within mRNA targets of the FMR1 protein family. b, Two
major RREs were inferred from FMRP iso1 and iso7 binding sites. c, Distribution of FMRP
binding sites, color-coded based on cERMIT-inferred RREs, across representative targets.
Open boxes and thick lines indicate CDS and UTRs, respectively; numbers indicate
nucleotide number.
Ascano et al. Page 10
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Figure 3. RNA binding assays using natural FMRP target sites containing ACUK and WGGA
RREs, and the effect of a KH2 mutation to its target RNA spectrum
a, EMSAs of RNAs representing UBE3A or PPP2CA binding sites containing various
RREs. Binding curves and constants are shown. The sequences of the RNAs are provided
with WGGA (yellow) and ACUK RREs (cyan) highlighted. b, Impact of KH2 mutation in
FMRP on target sites containing ACUK versus WGGA RREs. The RNA affinities of wt and
I304N FMRP iso1 were compared using binding sites in NF1 (ACUK) and FMR1 (WGGA).
c, Binding curves of wt and I304N FMRP for an RNA segment representing a mixed RRE
binding site in NF1, and several mutant sequence versions (ACUK (-), WGGA (-), and
ACUK, WGGA (-)). d, Comparison of FMRP iso1 affinity for RRE type in EMSAs and
FMRP iso1 and 7 wt and I304N PAR-CLIP libraries. Error bars in EMSA summary
represent s.d., n= 9 (ACUK), or 8 (WGGA). The ratio of sequence reads aligned to each
RRE binding site was calculated between wt and I304N FMRP PAR-CLIP libraries. The
average sequence-depth ratio of wt over I304N binding site, per RRE-type, are shown. Error
bars in the read-depth analyses represent the avg. min and max values across all subsampled
mutant libraries (n= 14 and 26 for iso 1 and 7, respectively).
Ascano et al. Page 11
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Figure 4. RRE-dependent enrichment criteria for FMRP association with mRNAs
a, RIP-chip experiments were performed using FLAG-HA-FMRP iso1. a-d, Cumulative
distribution fraction plots of FMRP targets based on indicated criteria. Transcripts were
grouped and color-coded based on indicated bins. Non-targets are mRNA transcripts with
zero PAR-CLIP binding sites, although detectable in the array; total is the sum of non-
targets and PAR-CLIP identified targets detectable by RIP-chip. d, Enrichment of ninety-
three PAR-CLIP identified ASD-related target genes. e, Immunoblot densitometry analysis
of top-ranking FMRP targets from RIP-chip and PAR-CLIP analyses in HEK293 and human
brain. In cell culture, target protein expression differences of indicated proteins were
determined upon induction of FMR1 iso1 or 7 expression. Similarly, relative protein
expression was measured using lysates prepared from indicated brain regions of four FXS
patients, compared to age/sex/anatomic-matched controls. Error bars represent s.e.m., with n
= 2-11 (depending on protein measured and whether the sample was HEK293 or brain
lysate). PABPC1 protein level served as loading and ratio control as it was a gene with
PAR-CLIP binding sites but showed no RIP-chip enrichment (-0.10 LFE).
Ascano et al. Page 12
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Figure 5. Ovarian phenotype in Fmr1-/- mice
Ovaries from wt and Fmr1-/- female mice were harvested at 3, 9, 12, and 18 wks and
processed for histological (a), morphological (b), and quantitative western analyses (c). a,
By 3 wks of age, histological staining (hematoxylin) of sectioned ovaries show greater than
expected number of follicles compared to wt. b, Ovaries from 18 wk old Fmr1-/- mice are
larger than wt and exhibit prominent cysts consistent with corpus luteal development. c,
Lysates were prepared from 9, 12, and 18 wk ovaries from two different wt and KO mice
each, and analysed by quantitative Western using Mtor, Sash1, and Tsc2 antibodies. As in
human samples, Pabpc1 was used for normalization.
Ascano et al. Page 13
Nature. Author manuscript; available in PMC 2013 June 20.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Supplementary resources (16)

... FMRP is a well-studied RNA-binding protein (RBP) that regulates local translation [8][9][10][11][12][13][14] and is involved in the control of calcium channels [15], actin cytoskeletal dynamics [16][17][18], chromatin dynamics [19], DNA damage response (DDR) [19,20], and replication stress response [21]. These cellular functions presume physical properties for FMRP, which are required for both the recognition and localization of messenger RNA (mRNA) targets and direct association with a multitude of proteins and protein complexes [22,23]. ...
... Please do not hesitate to contact the corresponding author if any additional information or clarification is required. 8 Disclosure Statement: The authors are unaware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the objectivity of this review. ...
Preprint
Full-text available
Silencing of the fragile X mental retardation 1 (FMR1) gene and consequently lack of synthesis of FMR protein (FMRP) are associated with fragile X syndrome, which is one of the most prevalent inherited intellectual disabilities. FMRP is a multifunctional protein involved in many cellular functions in nearly all subcellular compartments under normal conditions and under conditions of cellular stress in both neuronal and non-neuronal cell types. This is achieved through its trafficking signals, nuclear localization signal (NLS), nuclear export signal (NES), and nucleolar localization signal (NoLS), as well as its RNA and protein binding domains, and is modulated by various post-translational modifications such as phosphorylation, ubiquitination, sumoylation, and methylation. This review summarizes recent advances in understanding the interaction networks of FMRP with a special focus on FMRP stress-related functions, including stress granule formation, mitochondrion and endoplasmic reticulum plasticity, ribosome biogenesis, cell cycle control, and DNA damage response.
... It is a general regulator of RNA metabolism and especially of mRNA local translation in neurons (Banerjee et al., 2018). Its cognate mRNA targets are numerous and diverse, including mRNAs encoding proteins involved in neuronal plasticity like CamKIIα (calcium calmodulin-dependent kinase II) and cytoskeletal proteins like microtubule-associated protein 1B (MAP1B) (Ascano et al., 2012;Brown et al., 2001;Darnell et al., 2001;Maurin et al., 2018). Fmr1null mice are the murine model of FXS and have allowed characterization of numerous neurodevelopmental and plasticity defects consecutive to the absence of FMRP. ...
Article
Full-text available
The fragile X syndrome (FXS) represents the most prevalent form of inherited intellectual disability and is the first monogenic cause of autism spectrum disorder. FXS results from the absence of the RNA-binding protein FMRP (fragile X messenger ribonucleoprotein). Neuronal migration is an essential step of brain development allowing displacement of neurons from their germinal niches to their final integration site. The precise role of FMRP in neuronal migration remains largely unexplored. Using live imaging of postnatal rostral migratory stream (RMS) neurons in Fmr1 -null mice, we observed that the absence of FMRP leads to delayed neuronal migration and altered trajectory, associated with defects of centrosomal movement. RNA-interference-induced knockdown of Fmr1 shows that these migratory defects are cell-autonomous. Notably, the primary Fmrp mRNA target implicated in these migratory defects is microtubule-associated protein 1B (MAP1B). Knocking down MAP1B expression effectively rescued most of the observed migratory defects. Finally, we elucidate the molecular mechanisms at play by demonstrating that the absence of FMRP induces defects in the cage of microtubules surrounding the nucleus of migrating neurons, which is rescued by MAP1B knockdown. Our findings reveal a novel neurodevelopmental role for FMRP in collaboration with MAP1B, jointly orchestrating neuronal migration by influencing the microtubular cytoskeleton.
... FMRP is known to be expressed in the ovaries of f lies, mice, rats and humans [6,46], but there are not many studies delving into the exact function of FMRP during oogenesis. In humans, both males and females with FXS have been reported to be fertile and capable of reproduction [47,48]. ...
Article
Full-text available
Fragile X syndrome (FXS) is an inherited neurodevelopmental disorder and the leading genetic cause of autism spectrum disorders. FXS is caused by loss of function mutations in Fragile X mental retardation protein (FMRP), an RNA binding protein that is known to regulate translation of its target mRNAs, predominantly in the brain and gonads. The molecular mechanisms connecting FMRP function to neurodevelopmental phenotypes are well understood. However, neither the full extent of reproductive phenotypes, nor the underlying molecular mechanisms have been as yet determined. Here, we developed new fmr1 knockout zebrafish lines and show that they mimic key aspects of FXS neuronal phenotypes across both larval and adult stages. Results from the fmr1 knockout females also showed that altered gene expression in the brain, via the neuroendocrine pathway contribute to distinct abnormal phenotypes during ovarian development and oocyte maturation. We identified at least three mechanisms underpinning these defects, including altered neuroendocrine signaling in sexually mature females resulting in accelerated ovarian development, altered expression of germ cell and meiosis promoting genes at various stages during oocyte maturation, and finally a strong mitochondrial impairment in late stage oocytes from knockout females. Our findings have implications beyond FXS in the study of reproductive function and female infertility. Dissection of the translation control pathways during ovarian development using models like the knockout lines reported here may reveal novel approaches and targets for fertility treatments.
... FMRP is a well-studied RNA-binding protein (RBP) that regulates local translation [8][9][10][11][12][13][14] and controls calcium channels [15], actin cytoskeletal dynamics [16][17][18], chromatin dynamics [19], DNA damage response (DDR) [19,20], and replication stress response [21]. These cellular functions presume physical properties for FMRP, which are required for both the recognition and localization of messenger RNA (mRNA) targets and the direct association with a multitude of proteins and protein complexes [22,23]. ...
Article
Full-text available
Silencing of the fragile X messenger ribonucleoprotein 1 (FMR1) gene and a consequent lack of FMR protein (FMRP) synthesis are associated with fragile X syndrome, one of the most common inherited intellectual disabilities. FMRP is a multifunctional protein that is involved in many cellular functions in almost all subcellular compartments under both normal and cellular stress conditions in neuronal and non-neuronal cell types. This is achieved through its trafficking signals, nuclear localization signal (NLS), nuclear export signal (NES), and nucleolar localization signal (NoLS), as well as its RNA and protein binding domains, and it is modulated by various post-translational modifications such as phosphorylation, ubiquitination, sumoylation, and methylation. This review summarizes the recent advances in understanding the interaction networks of FMRP with a special focus on FMRP stress-related functions, including stress granule formation, mitochondrion and endoplasmic reticulum plasticity, ribosome biogenesis, cell cycle control, and DNA damage response.
... The increase in H 2 S in Fmr1 KOs could be due to upregulation of H 2 S synthetic enzymes: cystathionine βsynthase (CBS) and 3-mercaptopyruvate sulfur transferase (3-MST) (given that each is a known target of the translational repressor, FMRP [genetically silenced in the Fmr1 KOs]). 30 Furthermore, it is possible that the elevation in SQOR level and activity in this strain results as a consequence of increased H 2 S. Future work will need to determine the cause of H 2 S elevation and assess whether the effects of H 2 S on the pore are dependent on SQOR, since we did not directly assess the effect of H 2 S on the mPTP in this work. Despite these uncertainties, we were able to demonstrate that both inhibition and genetic silencing of SQOR induced mPTP opening in wild-type mouse heart mitochondria in vitro and in situ. ...
Article
Full-text available
Pathological opening of the mitochondrial permeability transition pore (mPTP) is implicated in the pathogenesis of many disease processes such as myocardial ischemia, traumatic brain injury, Alzheimer's disease, and diabetes. While we have gained insight into mPTP biology over the last several decades, the lack of translation of this knowledge into successful clinical therapies underscores the need for continued investigation and use of different approaches to identify novel regulators of the mPTP with the hope of elucidating new therapeutic targets. Although the mPTP is known to be a voltage‐gated channel, the identity of its voltage sensor remains unknown. Here we found decreased gating potential of the mPTP and increased expression and activity of sulfide quinone oxidoreductase (SQOR) in newborn Fragile X syndrome (FXS) mouse heart mitochondria, a model system of coenzyme Q excess and relatively decreased mPTP open probability. We further found that pharmacological inhibition and genetic silencing of SQOR increased mPTP open probability in vitro in adult murine cardiac mitochondria and in the isolated‐perfused heart, likely by interfering with voltage sensing. Thus, SQOR is proposed to contribute to voltage sensing by the mPTP and may be a component of the voltage sensing apparatus that modulates the gating potential of the mPTP.
Article
Objective The molecular mechanism of electroconvulsive therapy (ECT) for schizophrenia remains unclear. The aim of this study was to uncover the underlying biological mechanisms of ECT in the treatment of schizophrenia using a transcriptional dataset. Methods The peripheral blood mRNA sequencing data of eight patients (before and after ECT) and eight healthy controls were analyzed by integrated co-expression network analysis and the differentially expressed genes were analyzed by cluster analysis. Gene set overlap analysis was performed using the hypergeometric distribution of phypfunction in R. Associations of these gene sets with psychiatric disorders were explored. Tissue-specific enrichment analysis, gene ontology enrichment analysis, and protein–protein interaction enrichment analysis were used for gene set organization localization and pathway analysis. Results We found the genes of the green-yellow module were significantly associated with the effect of ECT treatment and the common gene variants of schizophrenia ( P = 0.0061; family-wise error correction). The genes of the green-yellow module are mainly enriched in brain tissue and mainly involved in the pathways of neurotrophin, mitogen-activated protein kinase and long-term potentiation. Conclusion Genes associated with the efficacy of ECT were predominantly enriched in neurotrophin, mitogen-activated protein kinase and long-term potentiation signaling pathways.
Preprint
Full-text available
Repetitive behaviors are core symptoms of autism spectrum disorders (ASD) and fragile X syndrome (FXS), the prevalent genetic cause of intellectual disability and autism. The nigrostriatal dopamine (DA) circuit rules movement and habit formation; therefore, its dysregulation stands as a leading substrate for repetitive behaviors. However, beyond indirect evidence, specific assessment of nigral DA neuron activity in ASD and FXS models is lacking. Here, we show that hyperactivity of substantia nigra pars compacta (SNpc) DA neurons is an early feature of FXS. The underlying mechanisms rely on mGluR1 and ErbB receptors. Up-regulation of ErbB4 and ErbB2 in nigral DA neurons drives neuronal hyperactivity and repetitive behaviors of the FXS mouse, simultaneously rescued by ErbB inhibition. In conclusion, beyond providing the first evidence of dysregulation of the SNpc DA nucleus in FXS, we identify novel targets - ErbB receptors - whose inhibition proficiently attenuates repetitive behaviors, thus opening an avenue toward innovative therapies for ASD and FXS.
Article
Male patients with fragile X syndrome lack FMRl pro-tein due to silencing of the FMRl gene by amplification of a CGG repeat and subsequent methylation of the promoter region. The absence of FMRl protein leads to mental retardation, aberrant behavior, and macroor-chidism. Hardly anything is known about the physiologi-cal function of FMR1 and the pathological mechanisms leading to these symptoms. Therefore, we designed a knockout model for the fragile X syndrome in mice. The knockout mice lack normal Fmrl protein and show mac-roorchidism, learning deficits, and hyperactivity. Consequently, this knockout mouse may serve as a valuable tool in the elucidation of the physiological role of FMR7 and the mechanisms involved in macroorchidism, abnormal behavior, and mental retardation.
Chapter
The Fragile X syndrome (FXS) is the most frequent form of inherited mental retardation and also considered a monogenic cause of Autism Spectrum Disorder. FXS symptoms include neurodevelopmental delay, anxiety, hyperactivity, and autistic-like behavior. The disease is due to mutations or loss of the Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein abundant in the brain and gonads, the two organs mainly affected in FXS patients. FMRP has multiple functions in RNA metabolism, including mRNA decay, dendritic targeting of mRNAs, and protein synthesis. In neurons lacking FMRP, a wide array of mRNAs encoding proteins involved in synaptic structure and function are altered. As a result of this complex dysregulation, in the absence of FMRP, spine morphology and functioning is impaired. Consistently, model organisms for the study of the syndrome recapitulate the phenotype observed in FXS patients, such as dendritic spine anomalies and defects in learning. Here, we review the fundamentals of genetic and clinical aspects of FXS, devoting a specific attention to ASD comorbidity and FXS-related diseases. We also review the current knowledge on FMRP functions through structural, molecular, and cellular findings. Finally, we discuss the neuroanatomical, electrophysiological, and behavioral defects caused by FMRP loss, as well as the current treatments able to partially revert some of the FXS abnormalities.
Article
The fragile X syndrome is the most frequent form of inherited mental retardation in humans with an incidence of 1 in 1250 males and 1 in 2500 females. The clinical syndrome includes moderate to severe mental retardation, autistic behavior, macroorchidism, and facial features, such as long face with mandibular prognathism and large, everted ears. The molecular basis for this disease is a large expansion of a triplet repeat (CGG){sub n} in the 5{prime} untranslated region of the FMR1 gene. Due to this large expansion of the CGG repeat, the promoter region becomes methylated and the FMR1 gene is subsequently silenced. Hardly anything is known about the physiologic function of FMR1 and the pathologic mechanisms leading to these symptoms. Since the FMR1 gene is highly conserved in the mouse, we used the mouse to design a knockout model for the fragile X syndrome. These knockout mice lacking Fmrp have normal litter size suggesting that FMR1 is not essential in human gametogenesis and embryonic development. The knockout mice show the abnormalities also seen in the affected organs of human patients. Mutant mice show a gradual development through time of macroorchidism. In the knockout mice we observed cognitive defects in the form of deficits in learning (as shown by the hidden platform Morris water maze task) and behavioral abnormalities such as increased exploratory behavior and hyperactivity. Therefore this knockout mouse may serve as a valuable tool in studying the role of FMR1 in the fragile X syndrome and may serve as a model to elucidate the mechanisms involved in macroorchidism, abnormal behavior, and mental retardation.
Article
Fragile X is the most common known inherited cause of intellectual disability and autism, and it typically results from transcriptional silencing of FMR1 and loss of the encoded protein, FMRP (fragile X mental retardation protein). FMRP is an mRNA-binding protein that functions at many synapses to inhibit local translation stimulated by metabotropic glutamate receptors (mGluRs) 1 and 5. Recent studies on the biology of FMRP and the signaling pathways downstream of mGluR1/5 have yielded deeper insight into how synaptic protein synthesis and plasticity are regulated by experience. This new knowledge has also suggested ways that altered signaling and synaptic function can be corrected in fragile X, and human clinical trials based on this information are under way.
Article
Fragile X syndrome Is the most frequent form of inherited mental retardation and Is associated with a fragile site at Xg27.3. We identified human YAC clones that span fragile X site-induced translocation breakpoints coincident with the fragile X site. A gene (FMR-1) was identified within 8 four cosmid contig of YAC DNA that expresses a 4.8 kb message in human brain. Within a 7.4 kb EcoFII genomic fragment, containing FMR-1 exonic sequences distal to a CpG island previously shown to be hypermethylated in fragile X patients, is a fragile X site-induced breakpoint cluster region that exhibits length variation in fragile X chromosomes. This fragment contains a lengthy CGG repeat that is 250 by distal of the CpG island and maps within a FMR-1 axon. Localization of the brain-expressed FMR-1 gene to this EcoRl fragment suggests the involvement of this gene in the phenotypic expression of the fragile X syndrome.
Book
The Fragile X syndrome (FXS) is the most frequent form of inherited mental retardation and also considered a monogenic cause of Autism Spectrum Disorder. FXS symptoms include neurodevelopmental delay, anxiety, hyperactivity, and autistic-like behavior. The disease is due to mutations or loss of the Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein abundant in the brain and gonads, the two organs mainly affected in FXS patients. FMRP has multiple functions in RNA metabolism, including mRNA decay, dendritic targeting of mRNAs, and protein synthesis. In neurons lacking FMRP, a wide array of mRNAs encoding proteins involved in synaptic structure and function are altered. As a result of this complex dysregulation, in the absence of FMRP, spine morphology and functioning is impaired. Consistently, model organisms for the study of the syndrome recapitulate the phenotype observed in FXS patients, such as dendritic spine anomalies and defects in learning. Here, we review the fundamentals of genetic and clinical aspects of FXS, devoting a specific attention to ASD comorbidity and FXS-related diseases. We also review the current knowledge on FMRP functions through structural, molecular, and cellular findings. Finally, we discuss the neuroanatomical, electrophysiological, and behavioral defects caused by FMRP loss, as well as the current treatments able to partially revert some of the FXS abnormalities.