Science - USA (2022-01-28)

(Antfer) #1
INSIGHTS | PERSPECTIVES

GRAPHIC: KELLIE HOLOSKI/

SCIENCE

science.org SCIENCE

By Rebecca A. Ihrie^1 and Elizabeth P. Henske^2

T

uberous sclerosis complex (TSC) is a
devastating disease characterized by
distinctive tumors of the skin (an-
giofibromas), brain [cortical tubers,
subependymal nodules (SENs), and
subependymal giant cell astrocy-
tomas (SEGAs)], heart (rhabdomyomas),
lungs [lymphangioleiomyomatosis (LAM)],
and kidney (angiomyolipomas and renal
cell carcinoma) ( 1 ). Neurologic manifesta-
tions can also include seizures and TSC-as-
sociated neuropsychiatric disorder, which
encompasses aggressive behaviors, autism

spectrum disorders, intellectual disability,
and psychiatric disorders. TSC is caused
by heterozygous germline mutations that
inactivate TSC1 or TSC2, which normally
inhibit mechanistic target of rapamycin
complex 1 (mTORC1). mTORC1 inhibitors
are used for the treatment of brain, lung,
and kidney tumors and seizures but do not
improve all symptoms, and uncertainty
persists about how brain tumorigenesis
begins. On page 401 of this issue, Eichmül-
ler et al. ( 2 ) identify a precursor cell that
generates cortical tuber–like and subep-
endymal tumor–like cells, increasing the

understanding of neurological manifesta-
tions of TSC.
The unmet clinical need for patients
with TSC is enormous, especially for the
complex spectrum of neurologic manifesta-
tions. Although many neurological features
can present soon after birth, whether these
features are due to altered prenatal cellular
development versus additional effects on
postnatal processes such as synaptic prun-
ing, white matter maturation, and brain cir-
cuit connectivity remains unclear. A barrier
to progress in this area has been challenges
in the development of models that fully re-
capitulate the brain malformations and neu-

ropsychiatric manifestations of TSC. Many
animal models diverge from what is seen in
patients—for example, requiring loss of addi-
tional tumor suppressor genes for tumor de-
velopment or exhibiting seizures but without
evident cortical tubers ( 3 – 7 ).
Uncertainty persists about the mecha-
nisms underlying brain hamartomas, in-
cluding SEN, SEGA, and tubers. Second-hit
genetic events, which cause loss of function
in the remaining wild-type copy of TSC1 or
TSC2, occur in virtually all TSC-associated
renal and lung tumors, which tend to pres-
ent later in life (see the figure). The second
hits lead to unrestrained mTORC1 activity
and are believed to be required for tumori-
genesis. Second hits in TSC1 or TSC2 have
been detected in many SENs and SEGAs
but not in most cortical tubers; whether

these events are required for pathogenesis
is unknown ( 8 ).
The cell of origin for several manifesta-
tions of TSC (including angiomyolipomas,
LAM, and brain hamartomas) is not well de-
fined. An additional knowledge gap relates to
how TSC is affected by factors such as sex,
immune infiltrate, modifying genes, diet, and
environmental exposures. Phenotypes and
severity are not strictly genotype dependent;
individuals with TSC in the same family can
have widely different clinical presentation.
These knowledge gaps are interrelated with
respect to the optimization of disease models
for preclinical studies.
Eichmüller et al. generate organoids
(cell cultures that form three-dimensional
masses) using induced pluripotent stem cells
(iPSCs) that are derived from TSC patients
and then differentiated into neural lineages.
These organoids recapitulate two manifes-
tations of TSC: proliferative lesions that re-
semble SENs or SEGAs and giant cells similar
to those in cortical tubers. Using single-cell
RNA-sequencing, the authors identify a pu-
tative shared cell of origin for both types of
lesion, called a c audal late interneuron pro-
genitor (CLIP) cell. CLIP cells express low
amounts of TSC2 and thus are hypothesized
to be more sensitive to the loss of one TSC2
allele. CLIP cells are transcriptionally separa-
ble from nearby progenitor populations that
have been shown to have increased mTORC1
activity and susceptibility to tumor formation
( 7 ). The authors argue that brain lesions ini-
tially consist of CLIP cells, but other cell types
appear during disease progression.
The discovery of CLIP cells, and the hy-
pothesis that they are the precursor of both
cortical tubers and subependymal tumors, is
potentially a breakthrough in understanding
the neurologic manifestations of TSC. These
findings contrast with other recent work in
human embryonic stem cells cultured into
cortical spheroids ( 9 ), which showed that
loss of both alleles of TSC1 or TSC2 was re-
quired for altered differentiation and the
emergence of dysmorphic cells, supporting
a two-hit model of tuber development. This
suggests that more than one mechanism can
lead to tubers, perhaps depending on the
developmental stage. Human iPSC-derived
kidney organoids have been found to develop
angiomyolipoma-like tumors, but only in the
setting of biallelic inactivation of TSC2, con-
firming that two hits are required ( 10 ). As or-

CANCER

Modeling tuberous sclerosis with organoids


S ingle-cell profiling reveals a different path for the development of brain lesions


Angio-
myolipoma

Lymphangio-
leiomyomatosis

X XX

Cortical tuber
Subependymal
nodule
Subependymal
giant cell
astrocytoma

CLIP
cells

CLIP-like
cells?

TSC1/2

(^1) Cell & Developmental Biology and Neurological Surgery,
Vanderbilt University School of Medicine, Nashville, TN,
USA.^2 Pulmonary and Critical Care Medicine, Brigham and
Women’s Hospital, Harvard Medical School, Boston, MA,
USA. Email: [email protected]
Tumorigenesis in tuberous sclerosis complex
Heterozygous germline inactivating mutations in tuberous sclerosis complex 1 (TSC1) and TSC2 result in
distinct tumors developing throughout life. Brain lesions present early and likely arise from caudal late
interneuron progenitor (CLIP) cells, whereas kidney and pulmonary tumors present later in life when a second-
hit inactivating mutation occurs in the wild-type TSC1/2 allele, possibly in CLIP-like cells.
382 28 JANUARY 2022 • VOL 375 ISSUE 6579

Free download pdf