Cells 12 02299
Cells 12 02299
Cells 12 02299
Review
Progerin, an Aberrant Spliced Form of Lamin A, Is a Potential
Therapeutic Target for HGPS
Bae-Hoon Kim 1 , Yeon-Ho Chung 1 , Tae-Gyun Woo 1 , So-Mi Kang 2 , Soyoung Park 2 and Bum-Joon Park 1,2, *
1 Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; [email protected] (B.-H.K.);
[email protected] (Y.-H.C.); [email protected] (T.-G.W.)
2 Department of Molecular Biology, College of Natural Science, Pusan National University,
Busan 46231, Republic of Korea; [email protected] (S.-M.K.); [email protected] (S.P.)
* Correspondence: [email protected]
terminal cysteine. Second, the Zmpste24 endopeptidase cleaves the last three amino acids
ofThird,
prelamin
the A. Third,
newly the newly
exposed exposed
farnesyl farnesyl
cysteine cysteine is carboxyl-methylated
is carboxyl-methylated by a
by a prenyl protein-
specific
prenyl methyltransferase.
protein-specific Finally, the endopeptidase
methyltransferase. Finally, theremoves 15 carboxyl-terminal
endopeptidase removes 15 amino
car-
acids from theamino
boxyl-terminal protein, resulting
acids in the
from the release
protein, of mature
resulting lamin
in the A. of mature lamin A.
release
Figure 1. A
Figure 1. schematic representation
A schematic of of
representation thethe
post-translational
post-translationalprocessing of of
processing lamin A and
lamin progerin
A and progerin
and the effect of progerin inhibition on cells. Spontaneous mutations in LMNA (c.
and the effect of progerin inhibition on cells. Spontaneous mutations in LMNA (c. 1824C>T)1824C>T) in in
eggs, sperm, embryos, or during aging cause alternative splicing of the LMNA gene, leading to
eggs, sperm, embryos, or during aging cause alternative splicing of the LMNA gene, leading to the the
accumulation of progerin in the nuclear layer. Consequently, the accumulation of progerin renders
accumulation of progerin in the nuclear layer. Consequently, the accumulation of progerin renders
the cell unhealthy in a mechanophysiological manner. Inhibiting progerin by several methods
the cell unhealthy in a mechanophysiological manner. Inhibiting progerin by several methods (small
(small molecules, ASOs, base editors) can restore damaged cells (See table 1 for detail).
molecules, ASOs, base editors) can restore damaged cells (See Table 1 for detail).
It has been questioned whether the post-translational processing steps of prelamin A
It has been questioned whether the post-translational processing steps of prelamin A
areare
essential
essential inin
targeting
targeting the protein
the proteintotothethenuclear
nuclear envelope
envelope [16]. Mice
[16]. Micethat could
that coulddirectly
directly
produce mature lamin A without going through the usual
produce mature lamin A without going through the usual prelamin A synthesis prelamin A synthesis and pro-
and
cessing steps were created. However, no detectable disease phenotype
processing steps were created. However, no detectable disease phenotype was observed was observed in in
thethe
mice
mice andandthethe
nuclear
nuclear membrane
membrane ofof
mature
mature lamin
laminAA appeared
appeared normal
normal [16], suggesting
[16], suggesting
that prelamin A processing is minimally important for the
that prelamin A processing is minimally important for the nuclear targeting nuclear targeting ofof
mature
mature
lamin A and is independent of lamin B in laboratory
lamin A and is independent of lamin B in laboratory mice. mice.
HGPS
HGPS belongs
belongs toto
a group
a group of of
diseases
diseases called laminopathies,
called laminopathies, in in
which
which mutations
mutations across
across
thethe
LMNA gene result in a wide range of overlapping disorders
LMNA gene result in a wide range of overlapping disorders [17]. Genetic mapping[17]. Genetic mapping of
theofgenome
the genomefrom from
patients elucidated
patients that a sporadic,
elucidated autosomal-dominant
that a sporadic, autosomal-dominantde novode point
novo
mutation, c.1824C>T
point mutation, (p.G608G)
c.1824C>T (NM_170707.3)
(p.G608G) in exonin
(NM_170707.3) 11exon
of the
11human LMNA LMNA
of the human gene, me-
gene,
diates abnormal
mediates abnormalalternative
alternativesplicing [9,10],
splicing which
[9,10], produces
which produces an an
abnormal
abnormal variant
variantprotein
protein
called
calledprogerin,
progerin, which
which is is
responsible
responsible forfor
this accelerated
this accelerated aging
aging disease
disease[18–20]
[18–20](Figure
(Figure1).1).
The current review describes the discovery of progerin as a
The current review describes the discovery of progerin as a causative agent causative agent ofof
HGPS
HGPS
and
and provides
provides evidence
evidence of of
itsits
deleterious
deleterious effects when
effects when expressed
expressed intracellularly,
intracellularly,andandthe
the
benefits of inhibiting its expression. Additionally, it introduces efforts to develop therapies
and clinical trials for HGPS.
Cells 2023, 12, 2299 3 of 17
into the abnormal interactions caused by progerin [51]. The evaluation of miRNA expres-
sion profiles in HGPS and normal fibroblasts revealed an enriched set of overexpressed
miRNAs belonging to the 14q32.2–14q32.3 miRNA cluster and showed that inducing their
overexpression in normal fibroblasts reduced cell proliferation and increased senescence,
whereas inhibiting them in HGPS fibroblasts alleviated proliferation defects and senes-
cence and reduced progerin accumulation [52]. Progerin overexpression induced notable
changes in miRNA expression and confirmed that has-miR-59 (miR-59) was markedly
upregulated in cells from patients with HGPS and in multiple tissues of an HGPS mouse
model (LmnaG609G/G609G ) [53].
In this report, we describe the relationship between progerin and cellular homeostasis,
and its role in inducing premature aging in most cells. Several groups have examined
progerin as a biomarker of aging and indicated that it may be one of the few known
biological indicators that initiate the aging process at a certain age [54–57]. Recently,
fibroblasts cultured from older individuals were shown to have nuclear features similar
to HGPS cells [54]. Notably, although these cells expressed progerin mRNA transcripts
at barely detectable levels [54], they contained several abnormal nuclei that were clearly
positive for progerin-specific antibodies after prolonged culture [58,59], indicating that
progerin can be expressed in normal cells. To further investigate the biological relationship
between progerin expression and aging in normal humans, Djabali et al. examined skin
biopsies from 150 unaffected individuals. They found that similar splicing events occurred
in vivo at low levels in the skin of individuals of all ages [60]. Although the mRNA
expression level of progerin is low, it accumulates with age in a subpopulation of skin
fibroblasts and terminally differentiated keratinocytes [60,61], suggesting that research on
HGPS may improve our knowledge of physiological aging.
b. Systemic effect of progerin or effect of progerin on tissues
Over the last 20 years, several types of mice have been developed as animal models to
investigate diverse aspects of HGPS as follows: the knock-out or transgenic mice affecting
the whole-body level are Zmpste24−/− [62,63], transgenic G608G BAC [64], LmnaG609G [65],
Apo−/− LmnaG609G/G609G [66], Ldlr−/− , and LmnaG609G/G609G [67]; the mice affecting spe-
cific tissues or cells are LmnaLCS/LCS SM22αCre [66], Apoe−/− LmnaLCS/LCS SM22αCre [66],
LmnaLCS/LCS Tie2Cre [68,69], Lmnaf/f ; TC [70], and Prog-Tg [71]—see reference [72] for
further details. The limited number of patients with HGPS worldwide renders it difficult
to conduct longitudinal studies and clinical trials, and there is also a scarcity of human
samples available for ex vivo analyses. Therefore, the use of animal models and their
derived cell lines has contributed to the understanding of progeria phenotypes and is
particularly important in developing therapeutic reagents for the disease. The current
section aims to examine how progerin affects the phenotypes of various cell types and
mouse models by compiling research carried out by various experts.
Blood vascular diseases are the predominant cause of death in classical HGPS [73].
As childhood progresses, other symptoms appear, including hair loss, joint stiffness, body
fat loss, osteoporosis, and other aspects of physiological aging. The most clinically rel-
evant aspect of HGPS is the hardening of the arteries (atherosclerosis), which leads to
premature death from cardiovascular disease or stroke at an average age of approximately
15 years [74]. In HGPS, atherosclerosis is accompanied by pathological changes in the aortic
wall, including severe vascular smooth muscle cell (VSMC) depletion in the media, extracel-
lular matrix deposition, calcification, and early thickening of the aortic wall [61,75,76]. Skin
tissue sections from patients with HGPS have shown that progerin accumulates primarily
in the nuclei of vascular cells, suggesting that its accumulation has a direct association with
vascular diseases in progeria [19]. Similarly, several reports have suggested that progerin
in vascular muscles could accelerate atherosclerosis by inducing endoplasmic reticulum
(ER) stress, DNA damage, wound healing impairment, mislocalization of a myocardin-
related transcription factor, and replication stress [77–81]. Recently, Hamczyk et al. (2018)
produced the first mouse model (Apoe–/– LmnaLCS/LCS SM22αCre) with progerin-induced
atherosclerosis acceleration expressing progerin specifically in VSMCs and demonstrated
Cells 2023, 12, 2299 6 of 17
ering the currently available information, adenine base editors (ABEs) appear to be more
advantageous than CRISPR/Cas9 approaches because they do not induce double-stranded
DNA breaks, inhibit lamin A, or efficiently correct mutations that cause HGPS. Furthermore,
it prevents HGPS-associated vascular features and extends the lifespan more than any other
tested treatment [108]. Recently, the transient expression of an ABE and single-guide RNA
using MS2 bacteriophage-lentivirus chimeric particles corrected the mutation in 20.8–24.1%
of skin cells in an HGPS mouse model (human tetop-LAG608G minigene), indicating
that it could be a good approach for future gene-editing therapies [109]. However, two
major challenges limit their application: moderate editing efficiency and the off-target
mutagenesis of DNA and RNA. Further pre-clinical studies are required to improve the
safety and effectiveness of ABE-based therapies by enhancing their efficiency, optimizing
vectors, fine-tuning doses, and defining the optimal treatment duration to achieve the best
outcomes for patients before moving to clinical trials.
aging features in both HGPS fibroblasts and model mice [120]. Pharmacological inhibition
of the NLRP3 inflammasome by the selective inhibitor MCC950 led to an improvement in
the cellular phenotype, a significant extension of lifespan in a mouse model (Zmpste24−/− ),
and a reduction in inflammasome-dependent inflammation [95]. MG132 was able to reduce
the TNF-α-induced inflammatory cytokine secretion of IL-1β, Il-6, TNF-α, IFN-γ, and TGF-β
in HGPS-like patient cells [116]. Several in vitro and in vivo studies have been conducted
to ameliorate bone and adipose tissue conditions in progeria. A mouse model (transgenic
mice, tetop-LAG608G+; Sp7-tTA+) with the osteoblast-and osteocyte-inducible expression
of progerin [121] was used to investigate the recovery from HGPS bone abnormalities by
silencing the mutation and the beneficial effect of treatment with resveratrol. Complete
silencing of the transgenic progerin expression normalized the bone morphology and min-
eralization, including improvements in the frequency of rib fractures and callus formation,
an increased number of osteocytes, and normalized dentinogenesis. However, despite these
positive findings, resveratrol treatment showed no beneficial effects [122]. Using an HGPS
mouse model (transgenic G608G BAC), Cubria et al. (2020) showed that treatment with
pravastatin and zoledronic acid significantly improved bone structure, mechanical properties,
and cartilage structure parameters, thereby improving the musculoskeletal phenotype of
the disease [89]. Progerin accumulation and high paracrine activation in adipocyte tissue
caused chronic inflammation and cellular senescence in a tetop-LAG608G+ mouse model. The
pro-inflammatory cytokines IL-1α, IL-1β, IL-6, IL-17α, interferon gamma (IFNγ), and TNF-α
were significantly higher than in controls [123]. Additionally, the loss of fat and fat deposits
has been observed in LmnaG609G/G609G mice [65,124]. Hartinger et al. [125] tested the effects
of bar (a JAK1/2 inhibitor and an anti-inflammatory agent) and a combination of bar and lon-
afarnib (a farnesyltransferase inhibitor) on adipogenesis using skin-derived precursors (SKPs).
Compared to mock-treated HGPS SKPs, bar and the combination of bar and lonafarnib treat-
ments improved the differentiation of HGPS SKPs into adipocytes and lipid droplet formation,
demonstrating the beneficial effect of combination treatment on adipogenesis in HGPS and
other lipodystrophies [125]. Recently, HGPS-associated vascular pathological features were
recovered by CRISPR/dCas9-activated Oct4 expression, which extended the lifespan of a
mouse model (transgenic G608G BAC) [126]. The miR-59 was markedly upregulated in HGPS
patient cells and multiple tissues in an HGPS mouse model (LmnaG609G/G609G ). Treatment
with AAV9-mediated anti-miR-59 reduced fibrosis in several organisms, alleviated epidermal
thinning and dermal fat loss, and extended the longevity of mouse models [53]. The efforts
discussed in this section for efficient HGPS treatment are summarized in Table 1.
Table 1. Cont.
Table 2. The list of the clinical trials for HGPS and related progeria up to date.
After the identification of the effects of zoledronate and pravastatin, a second clinical
trial was initiated using these two drugs (NCT00731016), followed by a tri-therapy clinical
trial combining lonafarnib, zoledronate, and pravastatin (NCT00879034 and NCT00916747).
Despite the lack of additional improvements in the tri-therapy compared to lonafarnib
alone, as reported in the results of the last clinical trial [136], researchers continued their
efforts to find the optimal conditions for the tri-therapy combination.
Targeting the farnesylation and methylation of progerin with chemical inhibitors
ameliorates some progerin-induced changes, but several questions persist concerning
the underlying mechanisms. All tested strategies aimed at inhibiting isoprenylcysteine
carboxyl-methyltransferase (ICMT) improved progeroid features. However, genetic inac-
tivation and treatment with a target inhibitor (C75) appeared to increase progerin levels
in cells [137]. The main disadvantage of blocking FTase and ICMT to treat HGPS is that
these endogenous enzymes target several other proteins in addition to progerin. Therefore,
changes in homeostatic farnesylation and methylation following FTase and ICMT inhibition
may have deleterious side effects that partially offset the positive outcome of reducing
the progerin-induced disease. The impact of mTOR signaling downstream of AKT should
also be assessed, as mTOR haploinsufficiency extends the lifespan of the HGPS mouse
model (transgenic G608G BAC) [138]. Lonafarnib and everolimus reduced pathology in an
iPSC-derived tissue-engineered blood vessel model [139]. Furthermore, the combination
of ICMT-targeting drugs with mTOR inhibitors such as everolimus, which are currently
being tested in HGPS trials alongside lonafarnib (NCT02579044), has the potential to yield
further positive effects.
Park et al. developed treatment strategies that targeted progerin more specifically us-
ing progerinin (or SLC-D011), an inhibitor drug that directly targets progerin, induces its
degradation, and finally disrupts the interaction between progerin and lamin A. The oral ad-
ministration of progerinin to an HGPS mouse model (LmnaG609G/G609G ) increased its lifespan
by approximately 50% [59,96,124]. Before studies on diseased states, randomized, double-
blind, placebo-controlled, single ascending dose (SAD) studies including food interactions
were conducted. This was followed by multiple ascending dose (MAD) studies to evaluate the
safety, tolerability, pharmacokinetics, and pharmacodynamic profiles of progerinin (SLC-D011)
in healthy volunteers (NCT04512963). To the best of our knowledge, this is the first in-human
study of progerinin. Additionally, progerinin had an excellent safety profile across all tested
doses or food conditions (up to a maximum dose of 2400 mg). The study subjects in the phase
I trial tolerated the drug well and confirmed an increase in exposure to progerinin under
different food conditions, with fasting showing the least exposure, followed by low-fat and
high-fat. The final enrollment included 63 healthy volunteers, with 47 subjects in the SAD
phase and 16 subjects in the MAD phase, at one site in the USA. The drug–drug interaction
potential was not clinically significant; therefore, dose adjustment was not necessary because
a phase I, open-labeled, fixed-sequence study was safely completed to assess the effects of a
CYP3A4 inhibitor (itraconazole) and a CYP3A4 inducer (phenytoin ER) on the single-dose
pharmacokinetics of progerinin in healthy volunteers.
6. Concluding Remarks
In this review, we introduce progerin as a pathogenic factor that induces HGPS
(Figure 1) and provide relevant evidence in support of this. Multiple efforts to understand
the causes of HGPS and develop treatments and clinical studies have been described; how-
ever, not all researchers’ contributions were included in this short review (Tables 1 and 2).
We might suggest that genome editing is the best option for the treatment of monogenic
diseases. However, for systemic diseases such as HGPS, there are at least two requirements:
(i) the delivery of the system to every cell in the body, and (ii) the demonstration of the
absence of off-target effects in all cells. Furthermore, there is an indispensable need to
strengthen the regulation of genotoxicity with regard to gene therapy in pediatric diseases.
Finally, we believe that it is appropriate to expedite efficacy-based approvals for patients
suffering from HGPS, as long as no toxicity risks are raised. We look forward to the upcom-
Cells 2023, 12, 2299 12 of 17
ing clinical trials for HGPS and progeroid laminopathies and hope to learn more about the
relationship between progerin and aging.
Author Contributions: Conceptualization, B.-H.K. and B.-J.P.; investigation, B.-H.K., Y.-H.C., T.-G.W.,
S.-M.K. and S.P.; writing—original draft preparation, B.-H.K.; writing—review and editing, Y.-H.C.,
T.-G.W., S.-M.K. and S.P.; supervision, B.-J.P. All authors have read and agreed to the published
version of the manuscript.
Funding: This work was supported by the Progeria Research Foundation (Grant #PRF 2019-75), a
National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (NRF-
2020R1A4A1019322), and the 2023 Regional Industry-Linked University Open-Lab Development
Support Program through the Commercialization Promotion Agency for R&D Outcomes (COMPA)
funded by the Ministry of Science and ICT (2023openlab(RnD)_02) to B.-J.P.
Conflicts of Interest: Bae-Hoon Kim, Yeon-Ho Chung, Tae-Gyun Woo, and Bum-Joon Park are
employees of PRG S&T Co., Ltd.
References
1. Gilford, H. Progeria: A form of senilism. Practitioner 1904, 73, 188–217.
2. Merideth, M.A.; Gordon, L.B.; Clauss, S.; Sachdev, V.; Smith, A.C.; Perry, M.B.; Brewer, C.C.; Zalewski, C.; Kim, H.J.; Solomon,
B.; et al. Phenotype and course of Hutchinson-Gilford progeria syndrome. N. Engl. J. Med. 2008, 358, 592–604. [CrossRef]
3. Burke, B.; Stewart, C.L. Life at the edge: The nuclear envelope and human disease. Nat. Rev. Mol. Cell Biol. 2002, 3, 575–585.
[CrossRef]
4. Kipling, D.; Davis, T.; Ostler, E.L.; Faragher, R.G. What can progeroid syndromes tell us about human aging? Science 2004,
305, 1426–1431. [CrossRef]
5. Miller, R.A. “Accelerated aging”: A primrose path to insight? Aging Cell 2004, 3, 47–51. [CrossRef] [PubMed]
6. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [CrossRef]
7. Gordon, L.B.; Rothman, F.G.; López-Otín, C.; Misteli, T. Progeria: A paradigm for translational medicine. Cell 2014, 156, 400–407.
[CrossRef]
8. Ahmed, M.S.; Ikram, S.; Bibi, N.; Mir, A. Hutchinson-Gilford progeria syndrome: A premature aging disease. Mol. Neurobiol.
2018, 55, 4417–4427. [CrossRef] [PubMed]
9. Eriksson, M.; Brown, W.T.; Gordon, L.B.; Glynn, M.W.; Singer, J.; Scott, L.; Erdos, M.R.; Robbins, C.M.; Moses, T.Y.; Berglund,
P.; et al. Recurrent de novo point mutations in lamin a cause Hutchinson-Gilford progeria syndrome. Nature 2003, 423, 293–298.
[CrossRef]
10. De Sandre-Giovannoli, A.; Bernard, R.; Cau, P.; Navarro, C.; Amiel, J.; Boccaccio, I.; Lyonnet, S.; Stewart, C.L.; Munnich, A.; Le
Merrer, M.; et al. Lamin a truncation in Hutchinson-Gilford progeria. Science 2003, 300, 2055. [CrossRef]
11. Lin, F.; Worman, H.J. Structural organization of the human gene encoding nuclear lamin a and nuclear lamin c. J. Biol. Chem.
1993, 268, 16321–16326. [CrossRef]
12. Wydner, K.L.; McNeil, J.A.; Lin, F.; Worman, H.J.; Lawrence, J.B. Chromosomal assignment of human nuclear envelope protein
genes LMNA, LMNB1, and LBR by fluorescence in situ hybridization. Genomics 1996, 32, 474–478. [CrossRef]
13. Fisher, D.Z.; Chaudhary, N.; Blobel, G. CDNA sequencing of nuclear lamins a and c reveals primary and secondary structural
homology to intermediate filament proteins. Proc. Natl. Acad. Sci. USA 1986, 83, 6450–6454. [CrossRef] [PubMed]
14. Mounkes, L.C.; Burke, B.; Stewart, C.L. The a-type lamins: Nuclear structural proteins as a focus for muscular dystrophy and
cardiovascular diseases. Trends Cardiovasc. Med. 2001, 11, 280–285. [CrossRef]
15. Davies, B.S.; Fong, L.G.; Yang, S.H.; Coffinier, C.; Young, S.G. The posttranslational processing of prelamin a and disease. Annu.
Rev. Genom. Hum. Genet. 2009, 10, 153–174. [CrossRef]
16. Coffinier, C.; Jung, H.J.; Li, Z.; Nobumori, C.; Yun, U.J.; Farber, E.A.; Davies, B.S.; Weinstein, M.M.; Yang, S.H.; Lammerding,
J.; et al. Direct synthesis of lamin a, bypassing prelamin a processing, causes misshapen nuclei in fibroblasts but no detectable
pathology in mice. J. Biol. Chem. 2010, 285, 20818–20826. [CrossRef] [PubMed]
17. Worman, H.J.; Bonne, G. “Laminopathies”: A wide spectrum of human diseases. Exp. Cell Res. 2007, 313, 2121–2133. [CrossRef]
18. Goldman, R.D.; Shumaker, D.K.; Erdos, M.R.; Eriksson, M.; Goldman, A.E.; Gordon, L.B.; Gruenbaum, Y.; Khuon, S.; Mendez,
M.; Varga, R.; et al. Accumulation of mutant lamin a causes progressive changes in nuclear architecture in Hutchinson-Gilford
progeria syndrome. Proc. Natl. Acad. Sci. USA 2004, 101, 8963–8968. [CrossRef]
19. McClintock, D.; Gordon, L.B.; Djabali, K. Hutchinson-Gilford progeria mutant lamin a primarily targets human vascular cells as
detected by an anti-lamin a g608g antibody. Proc. Natl. Acad. Sci. USA 2006, 103, 2154–2159. [CrossRef] [PubMed]
20. Kashyap, S.; Shanker, V.; Sharma, N. Hutchinson-Gilford progeria syndrome: A rare case report. Indian Dermatol. Online J. 2014,
5, 478–481. [CrossRef]
21. Nissan, X.; Blondel, S.; Navarro, C.; Maury, Y.; Denis, C.; Girard, M.; Martinat, C.; De Sandre-Giovannoli, A.; Levy, N.; Peschanski,
M. Unique preservation of neural cells in Hutchinson- Gilford progeria syndrome is due to the expression of the neural-specific
mir-9 microRNA. Cell Rep. 2012, 2, 1–9. [CrossRef] [PubMed]
Cells 2023, 12, 2299 13 of 17
22. Young, S.G.; Jung, H.J.; Lee, J.M.; Fong, L.G. Nuclear lamins and neurobiology. Mol. Cell Biol. 2014, 34, 2776–2785. [CrossRef]
23. Liu, G.H.; Barkho, B.Z.; Ruiz, S.; Diep, D.; Qu, J.; Yang, S.L.; Panopoulos, A.D.; Suzuki, K.; Kurian, L.; Walsh, C.; et al.
Recapitulation of premature ageing with iPSCs from Hutchinson-Gilford progeria syndrome. Nature 2011, 472, 221–225. [CrossRef]
24. Zhang, J.; Lian, Q.; Zhu, G.; Zhou, F.; Sui, L.; Tan, C.; Mutalif, R.A.; Navasankari, R.; Zhang, Y.; Tse, H.F.; et al. A human iPSC
model of Hutchinson Gilford progeria reveals vascular smooth muscle and mesenchymal stem cell defects. Cell Stem Cell 2011,
8, 31–45. [CrossRef] [PubMed]
25. Jung, H.J.; Coffinier, C.; Choe, Y.; Beigneux, A.P.; Davies, B.S.; Yang, S.H.; Barnes, R.H., 2nd; Hong, J.; Sun, T.; Pleasure, S.J.; et al.
Regulation of prelamin a but not lamin c by mir-9, a brain-specific microRNA. Proc. Natl. Acad. Sci. USA 2012, 109, E423–E431.
[CrossRef]
26. Jung, H.J.; Tu, Y.; Yang, S.H.; Tatar, A.; Nobumori, C.; Wu, D.; Young, S.G.; Fong, L.G. New LMNA knock-in mice provide a
molecular mechanism for the ‘segmental aging’ in Hutchinson-Gilford progeria syndrome. Hum. Mol. Genet. 2014, 23, 1506–1515.
[CrossRef]
27. Coffinier, C.; Chang, S.Y.; Nobumori, C.; Tu, Y.; Farber, E.A.; Toth, J.I.; Fong, L.G.; Young, S.G. Abnormal development of the
cerebral cortex and cerebellum in the setting of lamin b2 deficiency. Proc. Natl. Acad. Sci. USA 2010, 107, 5076–5081. [CrossRef]
28. Coffinier, C.; Fong, L.G.; Young, S.G. Lincing lamin b2 to neuronal migration: Growing evidence for cell-specific roles of b-type
lamins. Nucleus 2010, 1, 407–411. [CrossRef]
29. Coffinier, C.; Jung, H.J.; Nobumori, C.; Chang, S.; Tu, Y.; Barnes, R.H., 2nd; Yoshinaga, Y.; de Jong, P.J.; Vergnes, L.; Reue, K.; et al.
Deficiencies in lamin b1 and lamin b2 cause neurodevelopmental defects and distinct nuclear shape abnormalities in neurons.
Mol. Biol. Cell 2011, 22, 4683–4693. [CrossRef]
30. Padiath, Q.S.; Fu, Y.H. Autosomal dominant leukodystrophy caused by lamin b1 duplications a clinical and molecular case study
of altered nuclear function and disease. Methods Cell Biol. 2010, 98, 337–357. [CrossRef]
31. Padiath, Q.S.; Saigoh, K.; Schiffmann, R.; Asahara, H.; Yamada, T.; Koeppen, A.; Hogan, K.; Ptácek, L.J.; Fu, Y.H. Lamin b1
duplications cause autosomal dominant leukodystrophy. Nat. Genet. 2006, 38, 1114–1123. [CrossRef] [PubMed]
32. Dreesen, O.; Chojnowski, A.; Ong, P.F.; Zhao, T.Y.; Common, J.E.; Lunny, D.; Lane, E.B.; Lee, S.J.; Vardy, L.A.; Stewart, C.L.; et al.
Lamin b1 fluctuations have differential effects on cellular proliferation and senescence. J. Cell Biol. 2013, 200, 605–617. [CrossRef]
33. Cao, K.; Graziotto, J.J.; Blair, C.D.; Mazzulli, J.R.; Erdos, M.R.; Krainc, D.; Collins, F.S. Rapamycin reverses cellular phenotypes and
enhances mutant protein clearance in Hutchinson-Gilford progeria syndrome cells. Sci. Transl. Med. 2011, 3, 89ra58. [CrossRef]
34. Vidak, S.; Kubben, N.; Dechat, T.; Foisner, R. Proliferation of progeria cells is enhanced by lamina-associated polypeptide 2α
(LAP2α) through expression of extracellular matrix proteins. Genes Dev. 2015, 29, 2022–2036. [CrossRef]
35. Gordon, L.B.; Norris, W.; Hamren, S.; Goodson, R.; LeClair, J.; Massaro, J.; Lyass, A.; D’Agostino, R.B., Sr.; Tuminelli, K.; Kieran,
M.W.; et al. Plasma Progerin in patients with Hutchinson-Gilford progeria syndrome: Immunoassay development and clinical
evaluation. Circulation 2023, 147, 1734–1744. [CrossRef]
36. Kubben, N.; Zhang, W.; Wang, L.; Voss, T.C.; Yang, J.; Qu, J.; Liu, G.H.; Misteli, T. Repression of the antioxidant nrf2 pathway in
premature aging. Cell 2016, 165, 1361–1374. [CrossRef]
37. Kychygina, A.; Dall’Osto, M.; Allen, J.A.M.; Cadoret, J.C.; Piras, V.; Pickett, H.A.; Crabbe, L. Progerin impairs 3-d genome
organization and induces fragile telomeres by limiting the DNTP pools. Sci. Rep. 2021, 11, 13195. [CrossRef]
38. Kudlow, B.A.; Stanfel, M.N.; Burtner, C.R.; Johnston, E.D.; Kennedy, B.K. Suppression of proliferative defects associated with
processing-defective lamin a mutants by htert or inactivation of p53. Mol. Biol. Cell 2008, 19, 5238–5248. [CrossRef] [PubMed]
39. Benson, E.K.; Lee, S.W.; Aaronson, S.A. Role of Progerin-induced telomere dysfunction in HGPS premature cellular senescence. J.
Cell Sci. 2010, 123, 2605–2612. [CrossRef]
40. Chojnowski, A.; Ong, P.F.; Wong, E.S.; Lim, J.S.; Mutalif, R.A.; Navasankari, R.; Dutta, B.; Yang, H.; Liow, Y.Y.; Sze, S.K.; et al.
Progerin reduces lap2α-telomere association in Hutchinson-Gilford progeria. eLife 2015, 4, e07759. [CrossRef] [PubMed]
41. Noda, A.; Mishima, S.; Hirai, Y.; Hamasaki, K.; Landes, R.D.; Mitani, H.; Haga, K.; Kiyono, T.; Nakamura, N.; Kodama, Y. Progerin,
the protein responsible for the Hutchinson-Gilford progeria syndrome, increases the unrepaired DNA damages following
exposure to ionizing radiation. Genes Environ. 2015, 37, 13. [CrossRef] [PubMed]
42. Chojnowski, A.; Ong, P.F.; Foo, M.X.R.; Liebl, D.; Hor, L.P.; Stewart, C.L.; Dreesen, O. Heterochromatin loss as a determinant of
Progerin-induced DNA damage in Hutchinson-Gilford progeria. Aging Cell 2020, 19, e13108. [CrossRef] [PubMed]
43. Ron, D.; Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat. Rev. Mol. Cell Biol. 2007,
8, 519–529. [CrossRef]
44. Stiber, J.A.; Rosenberg, P.B. The role of store-operated calcium influx in skeletal muscle signaling. Cell Calcium 2011, 49, 341–349.
[CrossRef]
45. Wang, W.P.; Wang, J.Y.; Lin, W.H.; Kao, C.H.; Hung, M.C.; Teng, Y.C.; Tsai, T.F.; Chi, Y.H. Progerin in muscle leads to thermogenic
and metabolic defects via impaired calcium homeostasis. Aging Cell 2020, 19, e13090. [CrossRef]
46. Luo, X.; Jiang, X.; Li, J.; Bai, Y.; Li, Z.; Wei, P.; Sun, S.; Liang, Y.; Han, S.; Li, X.; et al. Insulin-like growth factor-1 attenuates
oxidative stress-induced hepatocyte premature senescence in liver fibrogenesis via regulating nuclear p53-Progerin interaction.
Cell Death Dis. 2019, 10, 451. [CrossRef] [PubMed]
47. Luo, X.; Bai, Y.; He, S.; Sun, S.; Jiang, X.; Yang, Z.; Lu, D.; Wei, P.; Liang, Y.; Peng, C.; et al. Sirtuin 1 ameliorates defenestration in
hepatic sinusoidal endothelial cells during liver fibrosis via inhibiting stress-induced premature senescence. Cell Prolif. 2021,
54, e12991. [CrossRef]
Cells 2023, 12, 2299 14 of 17
48. Li, Y.; Jiang, X.; Zhang, Y.; Gao, Z.; Liu, Y.; Hu, J.; Hu, X.; Li, L.; Shi, J.; Gao, N. Nuclear accumulation of ubc9 contributes to
SUMOylation of lamin a/c and nucleophagy in response to DNA damage. J. Exp. Clin. Cancer Res. 2019, 38, 67. [CrossRef]
[PubMed]
49. Lu, X.; Djabali, K. Autophagic removal of farnesylated carboxy-terminal lamin peptides. Cells 2018, 7, 33. [CrossRef]
50. Bai, Y.; Liu, J.; Jiang, X.; Li, X.; Zhang, B.; Luo, X. Nucleophagic degradation of Progerin ameliorates defenestration in liver
sinusoidal endothelium due to sirt1-mediated deacetylation of nuclear lc3. Cells 2022, 11, 3918. [CrossRef]
51. Ahn, J.; Lee, J.; Jeong, S.; Jo, I.; Kang, S.M.; Park, B.J.; Ha, N.C. Structural basis for the interaction between unfarnesylated progerin
and the IG-like domain of lamin a/c in premature aging disorders. Biochem. Biophys. Res. Commun. 2022, 637, 210–217. [CrossRef]
52. Frankel, D.; Delecourt, V.; Novoa-Del-Toro, E.M.; Robin, J.D.; Airault, C.; Bartoli, C.; Carabalona, A.; Perrin, S.; Mazaleyrat, K.; De
Sandre-Giovannoli, A.; et al. Mir-376a-3p and mir-376b-3p overexpression in Hutchinson-Gilford progeria fibroblasts inhibits cell
proliferation and induces premature senescence. iScience 2022, 25, 103757. [CrossRef] [PubMed]
53. Hu, Q.; Zhang, N.; Sui, T.; Li, G.; Wang, Z.; Liu, M.; Zhu, X.; Huang, B.; Lu, J.; Li, Z.; et al. Anti-hsa-mir-59 alleviates premature
senescence associated with Hutchinson-Gilford progeria syndrome in mice. EMBO J. 2023, 42, e110937. [CrossRef] [PubMed]
54. Scaffidi, P.; Misteli, T. Lamin a-dependent nuclear defects in human aging. Science 2006, 312, 1059–1063. [CrossRef] [PubMed]
55. Skoczyńska, A.; Budzisz, E.; Dana, A.; Rotsztejn, H. New look at the role of Progerin in skin aging. Prz. Menopauzalny 2015,
14, 53–58. [CrossRef] [PubMed]
56. Viteri, G.; Chung, Y.W.; Stadtman, E.R. Effect of Progerin on the accumulation of oxidized proteins in fibroblasts from Hutchinson
Gilford progeria patients. Mech. Ageing Dev. 2010, 131, 2–8. [CrossRef] [PubMed]
57. Scaffidi, P.; Misteli, T. Lamin a-dependent misregulation of adult stem cells associated with accelerated ageing. Nat. Cell Biol.
2008, 10, 452–459. [CrossRef]
58. Cao, K.; Capell, B.C.; Erdos, M.R.; Djabali, K.; Collins, F.S. A lamin a protein isoform overexpressed in Hutchinson-Gilford
progeria syndrome interferes with mitosis in progeria and normal cells. Proc. Natl. Acad. Sci. USA 2007, 104, 4949–4954. [CrossRef]
[PubMed]
59. Lee, S.J.; Jung, Y.S.; Yoon, M.H.; Kang, S.M.; Oh, A.Y.; Lee, J.H.; Jun, S.Y.; Woo, T.G.; Chun, H.Y.; Kim, S.K.; et al. Interruption of
Progerin-lamin a/c binding ameliorates Hutchinson-Gilford progeria syndrome phenotype. J. Clin. Investig. 2016, 126, 3879–3893.
[CrossRef] [PubMed]
60. McClintock, D.; Ratner, D.; Lokuge, M.; Owens, D.M.; Gordon, L.B.; Collins, F.S.; Djabali, K. The mutant form of lamin a
that causes Hutchinson-Gilford progeria is a biomarker of cellular aging in human skin. PLoS ONE 2007, 2, e1269. [CrossRef]
[PubMed]
61. Olive, M.; Harten, I.; Mitchell, R.; Beers, J.K.; Djabali, K.; Cao, K.; Erdos, M.R.; Blair, C.; Funke, B.; Smoot, L.; et al. Cardiovascular
pathology in Hutchinson-Gilford progeria: Correlation with the vascular pathology of aging. Arter. Thromb. Vasc. Biol. 2010,
30, 2301–2309. [CrossRef]
62. Leung, G.K.; Schmidt, W.K.; Bergo, M.O.; Gavino, B.; Wong, D.H.; Tam, A.; Ashby, M.N.; Michaelis, S.; Young, S.G. Biochemical
studies of zmpste24-deficient mice. J. Biol. Chem. 2001, 276, 29051–29058. [CrossRef] [PubMed]
63. Pendás, A.M.; Zhou, Z.; Cadiñanos, J.; Freije, J.M.; Wang, J.; Hultenby, K.; Astudillo, A.; Wernerson, A.; Rodríguez, F.; Tryggvason,
K.; et al. Defective prelamin a processing and muscular and adipocyte alterations in zmpste24 metalloproteinase-deficient mice.
Nat. Genet. 2002, 31, 94–99. [CrossRef]
64. Varga, R.; Eriksson, M.; Erdos, M.R.; Olive, M.; Harten, I.; Kolodgie, F.; Capell, B.C.; Cheng, J.; Faddah, D.; Perkins, S.; et al.
Progressive vascular smooth muscle cell defects in a mouse model of Hutchinson-Gilford progeria syndrome. Proc. Natl. Acad.
Sci. USA 2006, 103, 3250–3255. [CrossRef]
65. Osorio, F.G.; Navarro, C.L.; Cadiñanos, J.; López-Mejía, I.C.; Quirós, P.M.; Bartoli, C.; Rivera, J.; Tazi, J.; Guzmán, G.; Varela, I.; et al.
Splicing-directed therapy in a new mouse model of human accelerated aging. Sci. Transl. Med. 2011, 3, 106ra107. [CrossRef]
66. Hamczyk, M.R.; Villa-Bellosta, R.; Gonzalo, P.; Andrés-Manzano, M.J.; Nogales, P.; Bentzon, J.F.; López-Otín, C.; Andrés, V.
Vascular smooth muscle-specific Progerin expression accelerates atherosclerosis and death in a mouse model of Hutchinson-
Gilford progeria syndrome. Circulation 2018, 138, 266–282. [CrossRef]
67. Nevado, R.M.; Hamczyk, M.R.; Gonzalo, P.; Andrés-Manzano, M.J.; Andrés, V. Premature vascular aging with features of plaque
vulnerability in an atheroprone mouse model of Hutchinson-Gilford progeria syndrome with LDLR deficiency. Cells 2020, 9, 2252.
[CrossRef]
68. Del Campo, L.; Sánchez-López, A.; González-Gómez, C.; Andrés-Manzano, M.J.; Dorado, B.; Andrés, V. Vascular smooth muscle
cell-specific progerin expression provokes contractile impairment in a mouse model of Hutchinson-Gilford progeria syndrome
that is ameliorated by nitrite treatment. Cells 2020, 9, 656. [CrossRef] [PubMed]
69. Del Campo, L.; Sánchez-López, A.; Salaices, M.; von Kleeck, R.A.; Expósito, E.; González-Gómez, C.; Cussó, L.; Guzmán-Martínez,
G.; Ruiz-Cabello, J.; Desco, M.; et al. Vascular smooth muscle cell-specific progerin expression in a mouse model of Hutchinson-
Gilford progeria syndrome promotes arterial stiffness: Therapeutic effect of dietary nitrite. Aging Cell 2019, 18, e12936. [CrossRef]
[PubMed]
70. Sun, S.; Qin, W.; Tang, X.; Meng, Y.; Hu, W.; Zhang, S.; Qian, M.; Liu, Z.; Cao, X.; Pang, Q.; et al. Vascular endothelium-targeted
sirt7 gene therapy rejuvenates blood vessels and extends life span in a Hutchinson-Gilford progeria model. Sci. Adv. 2020, 6,
eaay5556. [CrossRef]
Cells 2023, 12, 2299 15 of 17
71. Osmanagic-Myers, S.; Kiss, A.; Manakanatas, C.; Hamza, O.; Sedlmayer, F.; Szabo, P.L.; Fischer, I.; Fichtinger, P.; Podesser, B.K.;
Eriksson, M.; et al. Endothelial progerin expression causes cardiovascular pathology through an impaired mechanoresponse.
J. Clin. Investig. 2019, 129, 531–545. [CrossRef] [PubMed]
72. Benedicto, I.; Dorado, B.; Andrés, V. Molecular and cellular mechanisms driving cardiovascular disease in Hutchinson-Gilford
progeria syndrome: Lessons learned from animal models. Cells 2021, 10, 1157. [CrossRef] [PubMed]
73. Gordon, L.B.; Shappell, H.; Massaro, J.; D’Agostino, R.B., Sr.; Brazier, J.; Campbell, S.E.; Kleinman, M.E.; Kieran, M.W. Association
of lonafarnib treatment vs no treatment with mortality rate in patients with Hutchinson-Gilford progeria syndrome. JAMA 2018,
319, 1687–1695. [CrossRef] [PubMed]
74. Ullrich, N.J.; Gordon, L.B. Hutchinson-Gilford progeria syndrome. Handb. Clin. Neurol. 2015, 132, 249–264. [CrossRef]
75. Stehbens, W.E.; Wakefield, S.J.; Gilbert-Barness, E.; Olson, R.E.; Ackerman, J. Histological and ultrastructural features of
atherosclerosis in progeria. Cardiovasc. Pathol. 1999, 8, 29–39. [CrossRef]
76. Stehbens, W.E.; Delahunt, B.; Shozawa, T.; Gilbert-Barness, E. Smooth muscle cell depletion and collagen types in progeric arteries.
Cardiovasc. Pathol. 2001, 10, 133–136. [CrossRef]
77. Hamczyk, M.R.; Villa-Bellosta, R.; Quesada, V.; Gonzalo, P.; Vidak, S.; Nevado, R.M.; Andrés-Manzano, M.J.; Misteli, T.; López-
Otín, C.; Andrés, V. Progerin accelerates atherosclerosis by inducing endoplasmic reticulum stress in vascular smooth muscle
cells. EMBO Mol. Med. 2019, 11, e9736. [CrossRef]
78. Kinoshita, D.; Nagasawa, A.; Shimizu, I.; Ito, T.K.; Yoshida, Y.; Tsuchida, M.; Iwama, A.; Hayano, T.; Minamino, T. Progerin
impairs vascular smooth muscle cell growth via the DNA damage response pathway. Oncotarget 2017, 8, 34045–34056. [CrossRef]
79. Jiang, Y.; Ji, J.Y. Progerin-induced impairment in wound healing and proliferation in vascular endothelial cells. Front. Aging 2022,
3, 844885. [CrossRef]
80. Coll-Bonfill, N.; Mahajan, U.; Shashkova, E.V.; Lin, C.J.; Mecham, R.P.; Gonzalo, S. Progerin induces a phenotypic switch in
vascular smooth muscle cells and triggers replication stress and an aging-associated secretory signature. GeroScience 2023,
45, 965–982. [CrossRef]
81. von Kleeck, R.; Castagnino, P.; Assoian, R.K. Progerin mislocalizes myocardin-related transcription factor in Hutchinson-guilford
progeria syndrome. Vasc. Biol. 2022, 4, 1–10. [CrossRef] [PubMed]
82. Sánchez-López, A.; Espinós-Estévez, C.; González-Gómez, C.; Gonzalo, P.; Andrés-Manzano, M.J.; Fanjul, V.; Riquelme-Borja,
R.; Hamczyk, M.R.; Macías, Á.; Del Campo, L.; et al. Cardiovascular Progerin suppression and lamin a restoration rescue
Hutchinson-Gilford progeria syndrome. Circulation 2021, 144, 1777–1794. [CrossRef] [PubMed]
83. Boguslavsky, R.L.; Stewart, C.L.; Worman, H.J. Nuclear lamin a inhibits adipocyte differentiation: Implications for Dunnigan-type
familial partial lipodystrophy. Hum. Mol. Genet. 2006, 15, 653–663. [CrossRef]
84. Bidault, G.; Vatier, C.; Capeau, J.; Vigouroux, C.; Béréziat, V. Lmna-linked lipodystrophies: From altered fat distribution to cellular
alterations. Biochem. Soc. Trans. 2011, 39, 1752–1757. [CrossRef] [PubMed]
85. Najdi, F.; Krüger, P.; Djabali, K. Impact of progerin expression on adipogenesis in Hutchinson-Gilford progeria skin-derived
precursor cells. Cells 2021, 10, 1598. [CrossRef]
86. Xiong, Z.M.; LaDana, C.; Wu, D.; Cao, K. An inhibitory role of progerin in the gene induction network of adipocyte differentiation
from IPS cells. Aging 2013, 5, 288–303. [CrossRef]
87. Mateos, J.; Landeira-Abia, A.; Fafián-Labora, J.A.; Fernández-Pernas, P.; Lesende-Rodríguez, I.; Fernández-Puente, P.; Fernández-
Moreno, M.; Delmiro, A.; Martín, M.A.; Blanco, F.J.; et al. Itraq-based analysis of Progerin expression reveals mitochondrial
dysfunction, reactive oxygen species accumulation and altered proteostasis. Stem Cell Res. Ther. 2015, 6, 119. [CrossRef]
88. Gordon, C.M.; Gordon, L.B.; Snyder, B.D.; Nazarian, A.; Quinn, N.; Huh, S.; Giobbie-Hurder, A.; Neuberg, D.; Cleveland, R.;
Kleinman, M.; et al. Hutchinson-Gilford progeria is a skeletal dysplasia. J. Bone Miner. Res. 2011, 26, 1670–1679. [CrossRef]
89. Cubria, M.B.; Suarez, S.; Masoudi, A.; Oftadeh, R.; Kamalapathy, P.; DuBose, A.; Erdos, M.R.; Cabral, W.A.; Karim, L.; Collins,
F.S.; et al. Evaluation of musculoskeletal phenotype of the g608g progeria mouse model with lonafarnib, pravastatin, and
zoledronic acid as treatment groups. Proc. Natl. Acad. Sci. USA 2020, 117, 12029–12040. [CrossRef]
90. Gargiuli, C.; Schena, E.; Mattioli, E.; Columbaro, M.; D’Apice, M.R.; Novelli, G.; Greggi, T.; Lattanzi, G. Lamins and bone
disorders: Current understanding and perspectives. Oncotarget 2018, 9, 22817–22831. [CrossRef]
91. Bergo, M.O.; Gavino, B.; Ross, J.; Schmidt, W.K.; Hong, C.; Kendall, L.V.; Mohr, A.; Meta, M.; Genant, H.; Jiang, Y.; et al. Zmpste24
deficiency in mice causes spontaneous bone fractures, muscle weakness, and a prelamin A processing defect. Proc. Natl. Acad.
Sci. USA 2002, 99, 13049–13054. [CrossRef] [PubMed]
92. Bidault, G.; Garcia, M.; Capeau, J.; Morichon, R.; Vigouroux, C.; Béréziat, V. Progerin expression induces inflammation, oxidative
stress and senescence in human coronary endothelial cells. Cells 2020, 9, 1201. [CrossRef] [PubMed]
93. Kreienkamp, R.; Graziano, S.; Coll-Bonfill, N.; Bedia-Diaz, G.; Cybulla, E.; Vindigni, A.; Dorsett, D.; Kubben, N.; Batista, L.F.Z.;
Gonzalo, S. A cell-intrinsic interferon-like response links replication stress to cellular aging caused by Progerin. Cell Rep. 2018,
22, 2006–2015. [CrossRef]
94. Messner, M.; Ghadge, S.K.; Maurer, T.; Graber, M.; Staggl, S.; Christine Maier, S.; Pölzl, G.; Zaruba, M.M. ZMPSTE24 is associated
with elevated inflammation and Progerin mRNA. Cells 2020, 9, 1981. [CrossRef]
95. González-Dominguez, A.; Montañez, R.; Castejón-Vega, B.; Nuñez-Vasco, J.; Lendines-Cordero, D.; Wang, C.; Mbalaviele, G.;
Navarro-Pando, J.M.; Alcocer-Gómez, E.; Cordero, M.D. Inhibition of the NLRP3 inflammasome improves lifespan in animal
murine model of Hutchinson-Gilford progeria. EMBO Mol. Med. 2021, 13, e14012. [CrossRef]
Cells 2023, 12, 2299 16 of 17
96. Kang, S.M.; Yoon, M.H.; Lee, S.J.; Ahn, J.; Yi, S.A.; Nam, K.H.; Park, S.; Woo, T.G.; Cho, J.H.; Lee, J.; et al. Human WRN is an
intrinsic inhibitor of Progerin, abnormal splicing product of lamin a. Sci. Rep. 2021, 11, 9122. [CrossRef]
97. Huang, S.; Chen, L.; Libina, N.; Janes, J.; Martin, G.M.; Campisi, J.; Oshima, J. Correction of cellular phenotypes of Hutchinson-
Gilford progeria cells by RNA interference. Hum. Genet. 2005, 118, 444–450. [CrossRef]
98. Zamecnik, P.C.; Stephenson, M.L. Inhibition of Rous sarcoma virus replication and cell transformation by a specific oligodeoxynu-
cleotide. Proc. Natl. Acad. Sci. USA 1978, 75, 280–284. [CrossRef]
99. Bennett, C.F. Therapeutic antisense oligonucleotides are coming of age. Annu. Rev. Med. 2019, 70, 307–321. [CrossRef]
100. Scaffidi, P.; Misteli, T. Reversal of the cellular phenotype in the premature aging disease Hutchinson-Gilford progeria syndrome.
Nat. Med. 2005, 11, 440–445. [CrossRef]
101. Lee, J.M.; Nobumori, C.; Tu, Y.; Choi, C.; Yang, S.H.; Jung, H.J.; Vickers, T.A.; Rigo, F.; Bennett, C.F.; Young, S.G.; et al. Modulation
of lmna splicing as a strategy to treat prelamin a diseases. J. Clin. Investig. 2016, 126, 1592–1602. [CrossRef]
102. Abdelrahman, A.; Nielsen, M.W.; Stage, M.H.; Arnspang, E.C. Nuclear envelope morphology change upon repetitive treatment
with modified antisense oligonucleotides targeting Hutchinson-Gilford progeria syndrome. Biochem. Biophys. Rep. 2023,
33, 101411. [CrossRef] [PubMed]
103. Harhouri, K.; Navarro, C.; Baquerre, C.; Da Silva, N.; Bartoli, C.; Casey, F.; Mawuse, G.K.; Doubaj, Y.; Lévy, N.; De Sandre-
Giovannoli, A. Antisense-based Progerin downregulation in HGPS-like patients’ cells. Cells 2016, 5, 31. [CrossRef]
104. Fong, L.G.; Ng, J.K.; Lammerding, J.; Vickers, T.A.; Meta, M.; Coté, N.; Gavino, B.; Qiao, X.; Chang, S.Y.; Young, S.R.; et al.
Prelamin a and lamin a appear to be dispensable in the nuclear lamina. J. Clin. Investig. 2006, 116, 743–752. [CrossRef]
105. Puttaraju, M.; Jackson, M.; Klein, S.; Shilo, A.; Bennett, C.F.; Gordon, L.; Rigo, F.; Misteli, T. Systematic screening identifies
therapeutic antisense oligonucleotides for Hutchinson-Gilford progeria syndrome. Nat. Med. 2021, 27, 526–535. [CrossRef]
[PubMed]
106. Erdos, M.R.; Cabral, W.A.; Tavarez, U.L.; Cao, K.; Gvozdenovic-Jeremic, J.; Narisu, N.; Zerfas, P.M.; Crumley, S.; Boku, Y.; Hanson,
G.; et al. A targeted antisense therapeutic approach for Hutchinson-Gilford progeria syndrome. Nat. Med. 2021, 27, 536–545.
[CrossRef] [PubMed]
107. Gagliardi, M.; Ashizawa, A.T. The challenges and strategies of antisense oligonucleotide drug delivery. Biomedicines 2021, 9, 433.
[CrossRef]
108. Koblan, L.W.; Erdos, M.R.; Wilson, C.; Cabral, W.A.; Levy, J.M.; Xiong, Z.M.; Tavarez, U.L.; Davison, L.M.; Gete, Y.G.; Mao,
X.; et al. In vivo base editing rescues Hutchinson-Gilford progeria syndrome in mice. Nature 2021, 589, 608–614. [CrossRef]
109. Whisenant, D.; Lim, K.; Revêchon, G.; Yao, H.; Bergo, M.O.; Machtel, P.; Kim, J.S.; Eriksson, M. Transient expression of an adenine
base editor corrects the Hutchinson-Gilford progeria syndrome mutation and improves the skin phenotype in mice. Nat. Commun.
2022, 13, 3068. [CrossRef]
110. Clements, C.S.; Bikkul, M.U.; Ofosu, W.; Eskiw, C.; Tree, D.; Makarov, E.; Kill, I.R.; Bridger, J.M. Presence and distribution
of Progerin in HGPS cells is ameliorated by drugs that impact on the mevalonate and mtor pathways. Biogerontology 2019,
20, 337–358. [CrossRef] [PubMed]
111. Aveleira, C.A.; Ferreira-Marques, M.; Cortes, L.; Valero, J.; Pereira, D.; Pereira de Almeida, L.; Cavadas, C. Neuropeptide y
enhances Progerin clearance and ameliorates the senescent phenotype of human Hutchinson-Gilford progeria syndrome cells. J.
Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 1073–1078. [CrossRef] [PubMed]
112. Gabriel, D.; Roedl, D.; Gordon, L.B.; Djabali, K. Sulforaphane enhances Progerin clearance in Hutchinson-Gilford progeria
fibroblasts. Aging Cell 2015, 14, 78–91. [CrossRef] [PubMed]
113. Xu, X.; Wang, D.; Zheng, C.; Gao, B.; Fan, J.; Cheng, P.; Liu, B.; Yang, L.; Luo, Z. Progerin accumulation in nucleus pulposus cells
impairs mitochondrial function and induces intervertebral disc degeneration and therapeutic effects of sulforaphane. Theranostics
2019, 9, 2252–2267. [CrossRef]
114. Egesipe, A.L.; Blondel, S.; Lo Cicero, A.; Jaskowiak, A.L.; Navarro, C.; Sandre-Giovannoli, A.; Levy, N.; Peschanski, M.; Nissan, X.
Metformin decreases Progerin expression and alleviates pathological defects of Hutchinson-Gilford progeria syndrome cells. NPJ
Aging Mech. Dis. 2016, 2, 16026. [CrossRef]
115. Harhouri, K.; Navarro, C.; Depetris, D.; Mattei, M.G.; Nissan, X.; Cau, P.; De Sandre-Giovannoli, A.; Lévy, N. MG132-induced
Progerin clearance is mediated by autophagy activation and splicing regulation. EMBO Mol. Med. 2017, 9, 1294–1313. [CrossRef]
116. Harhouri, K.; Cau, P.; Casey, F.; Guedenon, K.M.; Doubaj, Y.; Van Maldergem, L.; Mejia-Baltodano, G.; Bartoli, C.; De Sandre-
Giovannoli, A.; Lévy, N. MG132 induces Progerin clearance and improves disease phenotypes in HGPS-like patients’ cells. Cells
2022, 11, 610. [CrossRef] [PubMed]
117. Zhang, N.; Hu, Q.; Sui, T.; Fu, L.; Zhang, X.; Wang, Y.; Zhu, X.; Huang, B.; Lu, J.; Li, Z. Unique Progerin C-terminal peptide
ameliorates Hutchinson-Gilford progeria syndrome phenotype by rescuing bubr1. Nat. Aging 2023, 3, 185–201. [CrossRef]
118. Vehns, E.; Arnold, R.; Djabali, K. Impact of MnTBAP and baricitinib treatment on Hutchinson-Gilford progeria fibroblasts.
Pharmaceuticals 2022, 15, 945. [CrossRef]
119. Monterrubio-Ledezma, F.; Navarro-García, F.; Massieu, L.; Mondragón-Flores, R.; Soto-Ponce, L.A.; Magaña, J.J.; Cisneros, B.
Rescue of Mitochondrial Function in Hutchinson-Gilford Progeria Syndrome by the Pharmacological Modulation of Exportin
CRM1. Cells 2023, 12, 275. [CrossRef]
120. Squarzoni, S.; Schena, E.; Sabatelli, P.; Mattioli, E.; Capanni, C.; Cenni, V.; D’Apice, M.R.; Andrenacci, D.; Sarli, G.; Pellegrino,
V.; et al. Interleukin-6 neutralization ameliorates symptoms in prematurely aged mice. Aging Cell 2021, 20, e13285. [CrossRef]
Cells 2023, 12, 2299 17 of 17
121. Schmidt, E.; Nilsson, O.; Koskela, A.; Tuukkanen, J.; Ohlsson, C.; Rozell, B.; Eriksson, M. Expression of the Hutchinson-Gilford
progeria mutation during osteoblast development results in loss of osteocytes, irregular mineralization, and poor biomechanical
properties. J. Biol. Chem. 2012, 287, 33512–33522. [CrossRef] [PubMed]
122. Strandgren, C.; Nasser, H.A.; McKenna, T.; Koskela, A.; Tuukkanen, J.; Ohlsson, C.; Rozell, B.; Eriksson, M. Transgene silencing of
the Hutchinson-Gilford progeria syndrome mutation results in a reversible bone phenotype, whereas resveratrol treatment does
not show overall beneficial effects. FASEB J. 2015, 29, 3193–3205. [CrossRef] [PubMed]
123. Revêchon, G.; Viceconte, N.; McKenna, T.; Sola Carvajal, A.; Vrtačnik, P.; Stenvinkel, P.; Lundgren, T.; Hultenby, K.; Franco, I.;
Eriksson, M. Rare progerin-expressing preadipocytes and adipocytes contribute to tissue depletion over time. Sci. Rep. 2017,
7, 4405. [CrossRef]
124. Kang, S.M.; Yoon, M.H.; Ahn, J.; Kim, J.E.; Kim, S.Y.; Kang, S.Y.; Joo, J.; Park, S.; Cho, J.H.; Woo, T.G.; et al. Progerinin, an
optimized progerin-lamin A binding inhibitor, ameliorates premature senescence phenotypes of Hutchinson-Gilford progeria
syndrome. Commun. Biol. 2021, 4, 5. [CrossRef] [PubMed]
125. Hartinger, R.; Lederer, E.M.; Schena, E.; Lattanzi, G.; Djabali, K. Impact of combined baricitinib and FTI treatment on adipogenesis
in Hutchinson-Gilford progeria syndrome and other lipodystrophic laminopathies. Cells 2023, 12, 1350. [CrossRef] [PubMed]
126. Kim, J.; Hwang, Y.; Kim, S.; Chang, Y.; Kim, Y.; Kwon, Y.; Kim, J. Transcriptional activation of endogenous oct4 via the crispr/dcas9
activator ameliorates Hutchinson-Gilford progeria syndrome in mice. Aging Cell 2023, 22, e13825. [CrossRef] [PubMed]
127. Capell, B.C.; Erdos, M.R.; Madigan, J.P.; Fiordalisi, J.J.; Varga, R.; Conneely, K.N.; Gordon, L.B.; Der, C.J.; Cox, A.D.; Collins, F.S.
Inhibiting farnesylation of Progerin prevents the characteristic nuclear blebbing of Hutchinson-Gilford progeria syndrome. Proc.
Natl. Acad. Sci. USA 2005, 102, 12879–12884. [CrossRef] [PubMed]
128. Toth, J.I.; Yang, S.H.; Qiao, X.; Beigneux, A.P.; Gelb, M.H.; Moulson, C.L.; Miner, J.H.; Young, S.G.; Fong, L.G. Blocking protein
farnesyltransferase improves nuclear shape in fibroblasts from humans with progeroid syndromes. Proc. Natl. Acad. Sci. USA
2005, 102, 12873–12878. [CrossRef] [PubMed]
129. Capell, B.C.; Olive, M.; Erdos, M.R.; Cao, K.; Faddah, D.A.; Tavarez, U.L.; Conneely, K.N.; Qu, X.; San, H.; Ganesh, S.K.; et al. A
farnesyltransferase inhibitor prevents both the onset and late progression of cardiovascular disease in a progeria mouse model.
Proc. Natl. Acad. Sci. USA 2008, 105, 15902–15907. [CrossRef]
130. Dhillon, S. Lonafarnib: First approval. Drugs 2021, 81, 283–289. [CrossRef] [PubMed]
131. Fong, L.G.; Frost, D.; Meta, M.; Qiao, X.; Yang, S.H.; Coffinier, C.; Young, S.G. A protein farnesyltransferase inhibitor ameliorates
disease in a mouse model of progeria. Science 2006, 311, 1621–1623. [CrossRef] [PubMed]
132. Verstraeten, V.L.; Peckham, L.A.; Olive, M.; Capell, B.C.; Collins, F.S.; Nabel, E.G.; Young, S.G.; Fong, L.G.; Lammerding, J. Protein
farnesylation inhibitors cause donut-shaped cell nuclei attributable to a centrosome separation defect. Proc. Natl. Acad. Sci. USA
2011, 108, 4997–5002. [CrossRef] [PubMed]
133. Blondel, S.; Egesipe, A.L.; Picardi, P.; Jaskowiak, A.L.; Notarnicola, M.; Ragot, J.; Tournois, J.; Le Corf, A.; Brinon, B.; Poydenot,
P.; et al. Drug screening on Hutchinson Gilford progeria pluripotent stem cells reveals aminopyrimidines as new modulators of
farnesylation. Cell Death Dis. 2016, 7, e2105. [CrossRef] [PubMed]
134. Basso, A.D.; Kirschmeier, P.; Bishop, W.R. Lipid posttranslational modifications. Farnesyl transferase inhibitors Thematic Review
Series. J. Lipid Res. 2006, 47, 15–31. [CrossRef]
135. Gordon, L.B.; Kleinman, M.E.; Miller, D.T.; Neuberg, D.S.; Giobbie-Hurder, A.; Gerhard-Herman, M.; Smoot, L.B.; Gordon, C.M.;
Cleveland, R.; Snyder, B.D.; et al. Clinical trial of a farnesyltransferase inhibitor in children with Hutchinson-Gilford progeria
syndrome. Proc. Natl. Acad. Sci. USA 2012, 109, 16666–16671. [CrossRef] [PubMed]
136. Gordon, L.B.; Kleinman, M.E.; Massaro, J.; D’Agostino, R.B., Sr.; Shappell, H.; Gerhard-Herman, M.; Smoot, L.B.; Gordon, C.M.;
Cleveland, R.H.; Nazarian, A.; et al. Clinical trial of the protein farnesylation inhibitors lonafarnib, pravastatin, and zoledronic
acid in children with Hutchinson-Gilford progeria syndrome. Circulation 2016, 134, 114–125. [CrossRef]
137. Chen, X.; Yao, H.; Kashif, M.; Revêchon, G.; Eriksson, M.; Hu, J.; Wang, T.; Liu, Y.; Tüksammel, E.; Strömblad, S.; et al.
A small-molecule ICMT inhibitor delays senescence of Hutchinson-Gilford progeria syndrome cells. eLife 2021, 10, e63284.
[CrossRef]
138. Cabral, W.A.; Tavarez, U.L.; Beeram, I.; Yeritsyan, D.; Boku, Y.D.; Eckhaus, M.A.; Nazarian, A.; Erdos, M.R.; Collins, F.S. Genetic
reduction of mtor extends lifespan in a mouse model of Hutchinson-Gilford progeria syndrome. Aging Cell 2021, 20, e13457.
[CrossRef]
139. Abutaleb, N.O.; Atchison, L.; Choi, L.; Bedapudi, A.; Shores, K.; Gete, Y.; Cao, K.; Truskey, G.A. Lonafarnib and everolimus
reduce pathology in IPSC-derived tissue engineered blood vessel model of Hutchinson-Gilford progeria syndrome. Sci. Rep.
2023, 13, 5032. [CrossRef]
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.