3841Circulating Nucleic Acids as Liquid Biopsies and Noninvasive Disease Biomarkers CHAPTER 490
p = 0.016
0.0
0.2
0.4
0.6
0.8
ACR grade ≥2
ACR grade 0
ACR grade 1
%ddcfDNA
p <0.001
0
1
2
3
AMR ≥2
AMR0
AMR1
%ddcfDNA
A B
p <0.001
0.0
0.2
0.6
0.8
None Moderate Severe
%ddcfDNA 0.4
Mild
C Severity of LVEF drop
FIGURE 490-5 Correlation of percentage of donor-derived cell-free DNA (%dd-cfDNA) measures with
heart transplant biopsy-graded rejection and allograft dysfunction by echocardiography. A. %dd-cfDNA
in relation to severity of acute cellular rejection (ACR) by histopathologic interpretation of endomyocardial
biopsy. Grade 0 rejection includes both ACR grade 0 and antibody-mediated rejection (AMR) grade 0.
P value obtained by generalized estimating equation approach comparing all categories. B. %dd-cfDNA in
relation to severity of AMR by histopathologic interpretation of the endomyocardial biopsy. Grade 0 rejection
includes both ACR grades 0 or 1 and AMR grade 0. P value obtained by generalized estimating equation
approach comparing all categories. C. %dd-cfDNA in relation to severity of allograft dysfunction measured
by echocardiography. Allograft dysfunction was defined as a reduction of left ventricular ejection fraction
(LVEF) by ≥5% and was further stratified by severity based on the magnitude of LVEF decline as no (<5%),
mild (5% to <10%), moderate (≥10% to <15%), or severe (≥15%) allograft dysfunction. P value obtained by
generalized estimating equation approach comparing all categories. (Reproduced with permission from
S Agbor-Enoh et al: Cell-free DNA to detect heart allograft acute rejection. Circulation 143:1184, 2021.)
Finally, the investigators demonstrated higher dd-cfDNA levels
associated with antibody-mediated rejection compared to cellular
(T-cell–mediated) rejection, as well as different cfDNA fragment
types and lengths during these two rejection processes, suggesting
that dd-cfDNA patterns could potentially help differentiate these two
rejection subtypes.
Parallel studies in lung, kidney, and liver transplantation have also
confirmed the utility of dd-cfDNA as a noninvasive acute rejection
biomarker.
A large, multicenter, prospective study in kidney transplantation (Diagnosing Acute Rejection in Kidney Transplant Recipients
[DART]) enrolled 384 patients, on whom 107 biopsies of the transplanted kidney were performed. The dd-cfDNA level discriminated
between biopsies showing any acute rejection and controls (no histologic rejection), with an AUC of 0.74, sensitivity of 59%, and specificity of 85% for acute rejection at a dd-cfDNA cutoff of ≥1.0%. Since
publication of the DART study, other investigations
of dd-cfDNA in kidney transplantation have shown
similar results, with notably high dd-cfDNA levels (median 1.4–2.9%) in patients with antibodymediated rejection and poorer diagnostic accuracy
for patients with early (Banff 1A or borderline)
T-cell–mediated rejection. Studies have also shown
elevated dd-cfDNA levels in patients who develop
donor-specific antibodies, even in the absence
of antibody-mediated rejection. In kidney transplantation, dd-cfDNA could be elevated due to
pathologies other than rejection (e.g., BK virus
nephropathy and pyelonephritis) and is more likely
a marker of severe graft injury rather than rejection
alone. Thus, follow-up testing to determine the
cause and nature of graft injury or use of dd-cfDNA
surveillance primarily in patients at high risk of
allograft rejection would be reasonable.
Studies in liver transplantation have consistently
demonstrated elevated dd-cfDNA levels during
episodes of biopsy-proven acute rejection. As in
kidney transplantation, levels appear to rise before
development of clinical manifestations of acute
rejection, with data demonstrating elevated levels
4–6 days before a rise in transaminase enzyme
levels and 8–15 days before biopsy confirmation of
rejection. It is important to note that steady-state
dd-cfDNA levels vary between transplanted organs,
likely relative to the organ’s mass, cellularity, or
vascularization, with levels of 5–10% seen in stable
liver transplant recipients and levels <0.1% seen in
heart transplant patients.
Similar patterns of dd-cfDNA have also been
seen after lung transplantation, with elevated levels
detected weeks prior to and at the time of transbronchial biopsy demonstrating acute rejection.
As in heart and kidney transplantation, dd-cfDNA
levels correlate with severity of the rejection event,
with greater discriminatory power seen for severe
rejection and higher levels seen in antibodymediated rejection compared to cellular (T-cell–
mediated) rejection.
Donor-derived cfDNA levels also appear to have
prognostic value, even in the absence of acute
allograft injury. In a lung transplant cohort, patients
with the highest average dd-cfDNA levels within
the first 3 months after transplant, in the absence of
rejection, were at high risk for subsequent chronic
allograft dysfunction and allograft failure. Similarly,
in kidney transplantation, donor-derived cfDNA
levels at hospital discharge were shown to predict
graft dysfunction at 1 year after transplant. As such,
average dd-cfDNA levels over time may reflect
cumulative graft injury and related long-term adverse outcomes.
Over the past few years, targeted next-generation sequencing assays
(AlloSure [CareDx, Inc.] and Prospera [Natera]) have been developed
to quantify dd-cfDNA levels after transplantation. These assays are
composed of panels of highly polymorphic SNPs that, when sequenced
and quantified, enable differentiation of donor and recipient cfDNA
molecules using bioinformatics approaches. Such assays do not require
donor or recipient genotyping, are rapid and relatively cost effective
(particularly when compared to whole genome sequencing), and are
currently available for clinical use.
In summary, dd-cfDNA can be reliably detected in the plasma of
transplant recipients. Levels of dd-cfDNA are high immediately after
transplantation but rapidly fall to a baseline level within 2 weeks, after
ischemia-reperfusion injury has subsided. These dd-cfDNA levels
then remain at stable low levels in the absence of graft injury, with
baseline levels varying by organ type, likely related to the mass of the
3842 PART 20 Frontiers
transplanted organ. Most studies performed to date have demonstrated
rising levels with development of acute rejection and other forms
of allograft injury, with highest discriminatory power for antibodymediated rejection and severe grades of acute cellular rejection, and
have shown that levels rapidly fall after successful rejection treatment.
dd-cfDNA testing cannot completely replace biopsies of the transplanted organ, which provide valuable immunohistochemical and
molecular diagnostic information that helps guide clinical treatment.
Rather, this noninvasive assay reduces the need for surveillance biopsies
and enables clinicians to selectively perform biopsies on patients for
whom they are truly indicated, with attendant improvements in safety,
cost, and patient satisfaction. The ultimate goal is to use dd-cfDNA to
“personalize” immunosuppressive management by enabling weaning of
therapy in stable patients with low dd-cfDNA levels and augmentation
of treatment in patients with rising levels suggestive of early graft injury.
■ MONITORING MICROBIAL DIVERSITY AND
INFECTION AFTER TRANSPLANTATION
Immunosuppressive therapies reduce the risk of allograft rejection
but increase the transplant recipient’s susceptibility to infectious complications. The balance between level of immune system suppression
and the competing risks of rejection and infection is delicate, and the
desirable “therapeutic window” for patient treatment is narrow. Furthermore, diagnosis of infectious complications after transplantation
is challenging, as patients are susceptible to a wide array of pathogens
and common diagnostic tests rely on an a priori hypothesis for the
source of infection and performance of specific antigen-based or PCRbased molecular tests. Hypothesis-free sequencing of the microbial
component of transplant recipients’ cfDNA offers a window into the
state of the immune response and potential diagnosis of infectious
complications.
In 2013, De Vlaminck et al performed shotgun sequencing of heart
and lung transplant recipient cfDNA followed by alignment to reference human and microbial databases. This work showed that 2% of
cfDNA sequences were microbial in origin, including viral, bacterial,
and fungal genomes. They then studied the microbiome composition
in plasma at different levels of taxonomic classification and showed
that viruses (73%) are more abundant than bacteria (25%) and fungi
(2%). Subsequent testing showed quantitative agreement between viral
counts as measured by sequencing and quantitative PCR assays for
common posttransplant viral pathogens such as herpesviruses.
Available data on clinical drug treatment and dosage, including calcineurin inhibitor use and antiviral prophylaxis, were used to analyze
drug-microbiome interactions. The investigators found that the structure of the virome is exquisitely sensitive to drug use and dosage—high
doses or levels of immunosuppressive drugs give rise to a virome
dominated by Anelloviridae or torque teno viruses, which are ubiquitous viruses that have no known pathogenic role in humans but have
been shown to replicate in the setting of immune system suppression.
Similarly, the relative amount of herpesviruses declines substantially
after introduction of the prophylactic antiviral agent valganciclovir
(Fig. 490-6). Using this approach, the investigators demonstrated dramatic changes in the viral composition of the microbiome in response
to introduction and weaning of immunosuppressive and antiviral therapies after transplantation.
Using a similar approach, De Vlaminck and colleagues analyzed
nonhuman cfDNA as a hypothesis-free approach to test for infectious
complications after lung transplantation. They showed that levels of
cytomegalovirus-derived sequences in cfDNA correlated well with
clinical testing results (AUC 0.91). They also identified adenovirus,
polyomavirus, herpesvirus, and microsporidia sequences in patients
with clinical infections that had eluded diagnosis, suggesting that
10%
3%
1%
60%
39% 31% 21%
30%
15%
34%
0–3
0–300 300–600
Antiviral drugs (Valganciclovir) (mg)
Immunosuppressant (Tacrolimus) ng/mL
600–900
3–6
6–9
9–12
12–15
72%
84%
14%
12%
54%
18%
80%
94%
2%
82%
4%
2%
87%
2%
82%
6%
75%
13%
87%
86%
Herpesvirales
Caudovirales
Adenoviridae
Anelloviridae
Polyomaviridae
Poxviridae
Retroviridae
Other
FIGURE 490-6 Relative viral genomic abundance as a function of drug dose in a cohort of heart and lung transplant recipients. Mean virome composition for patients
treated with the immunosuppressant tacrolimus (47 patients, 380 samples) as a function of antiviral drug dose (valganciclovir) and concentration of tacrolimus measured in
blood. To account for the delayed effect of the virome composition on drug dose, the data on drug doses were window average filtered (window size, 45 days). Herpesvirales
and caudovirales dominate the virome when patients receive low doses of immunosuppressants and antiviral drugs. Conversely, anelloviridae dominate the virome when
patients receive high doses of these drugs. (Reproduced with permission from I De Vlaminck et al: Temporal response of the human virome to immunosuppression and
antiviral therapy. Cell 155:1178, 2013.)
3843Circulating Nucleic Acids as Liquid Biopsies and Noninvasive Disease Biomarkers CHAPTER 490
Time period post transplant (months)
0
0
50
100
3 6 9 12 14 >15
Abundance (%)
Herpesvirales
Caudovirales
Adenoviridae
Anelloviridae
Polyomaviridae
Poxviridae
Retroviridae
Other
FIGURE 490-7 Viral genome abundance at the family and order level of taxonomic classification
for different time periods after heart and lung transplantation. The fraction of anelloviridae
expands rapidly in the first several months after transplantation. The fractions of herpesvirales,
caudovirales, and adenoviridae decrease in that same time period. After 6 months, the opposite
trends are observed. (Reproduced with permission from I De Vlaminck et al: Temporal response of
the human virome to immunosuppression and antiviral therapy. Cell 155:1178, 2013.)
No ACR Mild ACR Moderate
ACR
10
p = 0.003 p = 0.005
2
No CMV
A B
CMV
Infection
CMV
Disease
4
6
TTV DNA (log10 copies/mL)
8
10
2
4
6
TTV DNA (log10 copies/mL)
8
FIGURE 490-8 TTV (torque teno virus, anellovirus) load during (A) episodes of acute cellular rejection
(ACR) and (B) cytomegalovirus (CMV) infection and disease after liver transplantation. (Reproduced with
permission from P Ruiz et al: Torque teno virus is associated with the state of immune suppression early after
liver transplantation. Liver Transpl 25:302, 2019.)
cfDNA sequencing can be a powerful approach for nonbiased diagnosis of opportunistic infections after transplantation. However, several
obstacles to clinical implementation must first be overcome, including
the establishment of pathogen-specific thresholds to discriminate
between colonization, infection, and disease.
■ MONITORING HOST IMMUNITY
As mentioned previously, De Vlamink and colleagues demonstrated
that the Anelloviridae family of viruses predominated in the virome
of lung transplant recipients, as analyzed by plasma cfDNA sequencing, and accounted for 68% of the total viral population. The relative abundance of anellovirus cfDNA is exquisitely sensitive to
immunosuppression, ranging from <5% prior to transplant (e.g., in
the nonimmunosuppressed state) to 84% during
months 4.5–6 after transplantation, when immunosuppression is most profound. Starting at 6
months after transplantation, the relative amount
of anellovirus cfDNA declines, corresponding to
a reduction in intensity of immunosuppression
(Fig. 490-7).
The observation that anellovirus cfDNA levels rise and fall with degree of immune system
suppression gave rise to the hypothesis that the
abundance of this ubiquitous family of viruses
could be used as a marker of the overall strength
of the immune response. Indeed, a global marker
of the net state of immunosuppression has eluded
transplant physicians to date, who currently quantify doses and levels of individual immunosuppressive drugs but cannot reliably assess for their
effect or potency in combination. In support of
this hypothesis was the subsequent observation
that anellovirus load was lower (reflecting less
immunosuppression) in lung transplant recipients
who developed acute rejection and was relatively
high (indicating more profound immunosuppression) in patients who had a rejection-free
posttransplant course, suggesting that the anellovirus cfDNA load could be a surrogate marker of
immunocompetence.
Subsequent studies have confirmed these
observations after different solid organ transplant
procedures (Fig. 490-8). A prospective lung transplant
study showed a hazard ratio (HR) of 5.05 for infectious
complications and an HR of 0.48 for acute rejection with
every log10 increase in anellovirus load. Similar results
have been shown after liver and kidney transplantation,
with higher anellovirus levels associated with opportunistic infections and malignancy (sequelae of chronic
immunosuppression) and lower levels associated with
acute rejection and chronic allograft dysfunction. While
much work remains to be done in this arena, including
standardization of anellovirus assays and quantification,
prospective studies with well-defined sampling schedules
and endpoints, and improved understanding of genotype-specific effects on host immunity, the potential to
use anellovirus cfDNA load as a marker to titrate and
personalize immunosuppressive therapy may be within
reach.
In summary, cfDNA sequencing has myriad clinical
applications after solid organ transplantation. Donorderived cfDNA assays are currently available for acute
rejection monitoring and are being used for noninvasive
surveillance after heart, lung, and kidney transplantation.
These assays are highly sensitive, provide early detection of allograft injury, and can also be used to monitor
response to rejection therapy. cfDNA sequencing can
also be used to monitor changes in the microbiome after
transplantation, in response to alterations in immunosuppression and antimicrobial therapy, and can potentially be
used to diagnose opportunistic infections. Finally, quantification of
anellovirus cfDNA load offers the possibility of assessing the overall
status of the host immune response, which is a necessary first step for
providing individualized immunosuppressive therapy to transplant
recipients.
CELL-FREE DNA IN PRENATAL MEDICINE
Fetal genetic disorders are among the leading causes of stillbirth, neonatal death, and long-term developmental delay. Prenatal diagnosis
of fetal genetic conditions relies on invasive sampling of fetal tissues
via either amniocentesis (sampling of amniotic fluid) or chorionic
villus sampling (CVS; sampling of placental tissue). Prenatal diagnostic
3844 PART 20 Frontiers
tests are invasive, ultrasound-guided, needle-based procedures that
unfortunately carry a small risk of miscarriage, preterm rupture of the
membranes, and preterm birth. Historically, safer noninvasive screening for fetal genetic disorders, particularly aneuploidy (chromosomal
copy number abnormalities such as trisomy 21 or Down’s syndrome),
was performed by combining maternal serum analyte levels (e.g.,
pregnancy-associated plasma protein A [PAPP-A], β-human chorionic
gonadotropin [β-hCG], α-fetoprotein [AFP], inhibin) with sonographic assessment of the fetal nuchal translucency (a measurement of
the distance between the scalp and skin in the area of the fetal neck).
While traditional prenatal screening is safer than invasive diagnostic
tests, these modalities suffer from lower detection rates and higher
false-positive rates. Recent advances in cell-free fetal DNA technology
have introduced safe noninvasive prenatal tests with much higher
accuracy than traditional screening. This section describes the use of
cell-free fetal DNA testing in prenatal medicine.
■ NONINVASIVE PRENATAL TESTING
FOR FETAL ANEUPLOIDY
Passage of fetal cells into the maternal bloodstream was initially
described almost 80 years ago. A mismatch between fetal red blood
cells and maternal red blood cells was subsequently identified as the
cause of isoimmunization and hydrops fetalis, a potentially lethal
condition in which maternal antibodies develop against “foreign” fetal
red blood cell antigens. With the development of Rho(D) immune
globulin (RhoGAM) and its use to prevent RhD isoimmunization in
RhD-negative mothers carrying RhD-positive pregnancies, attention
then turned toward targeting fetal red blood cells for the detection of
fetal aneuploidy. This is because fetal red blood cells contain a nucleus
and genomic material, whereas maternal red blood cells do not. Unfortunately, initial research endeavors in the area failed to yield significant
results due to several inherent limitations. First, the number of fetal red
blood cells circulating among the more prevalent maternal red blood
cell background is quite small. Second, the half-life of the circulating
fetal red blood cells in maternal serum was found to be quite long,
and circulating cells may actually originate not just from the current
pregnancy but even from a prior pregnancy. Finally, the technology
available at the time, namely fluorescent-activated cell sorting, was
unable to accurately separate the cells to isolate the fetal cells from the
maternal background.
In 1990, Lo and colleagues determined that cell-free fetal DNA, and
not just fetal cells, crosses the placenta and could serve as a target for
fetal aneuploidy detection. This was subsequently confirmed when the
presence of DNA from the Y chromosome was found in the serum of
pregnant women carrying a male fetus. Over the next two decades,
cfDNA circulating in maternal blood was characterized, and different
genetic techniques were employed to delineate differences between
cell-free fetal DNA and cell-free maternal DNA. Cell-free fetal DNA
has several advantages as a diagnostic target over circulating fetal cells.
First, the fraction of fetal DNA is much higher (~5–20%) than the fraction of fetal cells in maternal circulation. Second, cell-free fetal DNA
declines within days of delivery, thereby reducing the risk of DNA from
previous pregnancies leading to erroneous results.
Unfortunately, in the case of fetal aneuploidy detection, the actual
sequence of fetal DNA from the chromosome of interest is identical
to the maternal DNA sequence, and the challenge is to distinguish one
from the other. In the early 2000s, the most promising technique for
distinguishing fetal from maternal DNA was leveraging epigenetic differences between the two, particularly in the chromosomes of interest,
namely chromosome 21 and chromosome 18. In 2008, two landmark
studies, one by Chiu and colleagues and one by Fan and colleagues,
described the utility of massive parallel sequencing technology as a
counting method for noninvasive detection of fetal aneuploidy. This
sequencing technology allowed for a comparison of the number of
sequencing reads from chromosome 21 against an expected reference
number of reads. In other words, if more sequences from chromosome
21 were found in maternal serum than what one would expect, the
fetus was suspected of having Down’s syndrome. While these findings
were based on analysis of limited cohorts, subsequent trials in more
robust and diverse cohorts confirmed these findings, and cfDNA testing became an approved clinical test in 2011.
While the initial focus of cfDNA testing was on pregnant women at
high risk for fetal aneuploidy (e.g., advanced maternal age, high-risk
status based on serum analyte screening, presence of fetal structural
anomalies), subsequent studies confirmed the utility of cfDNA technology in both high-risk and low-risk cohorts, opening the path for
widespread adoption. Today, the detection rate for the common aneuploidies (trisomies 21, 18, and 13) and for sex chromosome aneuploidy
using cfDNA testing is >98% (trisomy 21 detection as high as 99.8%),
and cfDNA testing is offered by both private companies and several
public nationwide screening programs. The positive predictive value of
these tests is variable and dependent on a priori maternal risk; therefore, most societal guidelines recommend confirmatory/diagnostic
testing using amniocentesis or CVS in cases of positive cfDNA testing.
■ NONINVASIVE PRENATAL SCREENING VERSUS
NONINVASIVE PRENATAL DIAGNOSIS VERSUS
NONINVASIVE PRENATAL TESTING
When data from clinical trials proved the improved performance of
cfDNA testing over traditional screening, it became unclear whether to
classify the test as noninvasive prenatal screening (NIPS) or noninvasive
prenatal diagnosis (NIPD). On the one hand, cfDNA testing leverages
circulating segments of fetal DNA in maternal serum, unlike traditional
screening, which is dependent on indirect analytes such as PAPP-A
or β-hCG. On the other hand, variable positive predictive values for
different aneuploidies still did not reach the diagnostic accuracy of
amniocentesis or CVS, and most cfDNA segments originate from the
placenta, rather than from the fetus. Therefore, over time, the label of
noninvasive prenatal testing (NIPT) became commonly used in both
the scientific literature and the lay literature. Today, much confusion
remains for both patients and providers as to the accuracy of these tests
and their inherent limitations.
■ DEPENDENCE ON FETAL FRACTION
As clinical experience with NIPT grew, it became clear that the accuracy of the test depends on the fraction of fetal DNA in maternal
serum. While fetal DNA can be found in maternal serum as early as
4–5 weeks’ gestation, the fetal fraction increases with gestational age,
and most NIPT programs recommend testing after 10 weeks’ gestation.
Although each NIPT company or national NIPT program uses a different fetal fraction cutoff to accurately report a result, a small percentage
of patients will receive a “no call” or uninterpretable result due to low
fetal fraction. Factors leading to “no call” also include early gestational
age (usually <10 weeks), maternal obesity, underlying maternal autoimmune disease or malignancy, and vanishing twin (twins in which
one fetus demises early in gestation). Several studies suggested an association between a no-call result and increased risk for aneuploidy, and
the appropriate management of a no-call result remains controversial,
with some recommending a repeat NIPT test, while others recommend
invasive diagnostic testing given the increased aneuploidy risk.
■ NIPT IN MULTIPLE GESTATIONS
While initial results questioned the utility of NIPT in multiple gestations, more recent reports from large cohorts suggest that the aneuploidy detection rate in twins mirrors that in singletons. Moreover,
many NIPT programs report zygosity in cases of multiple gestations.
This is particularly important since one would assume that NIPT
results from monozygotic twins (twins originating from a single
embryo) would indicate the presence of euploidy or aneuploidy in
both twins, and zygosity testing is also important when the number of
placentas has not been determined or remains unclear based on sonography. Monochorionic twins (all of whom are monozygotic twins)
are considered higher risk than dichorionic twins (most of whom are
dizygotic twins but can also be monozygotic twins) and are managed
differently. Newer NIPT platforms can also distinguish the presence of
aneuploidy in a single fetus in cases of dizygotic twins. NIPT is still not
validated for triplet gestations or multiple gestations in which a single
fetal reduction or fetal loss occurs.
3845Circulating Nucleic Acids as Liquid Biopsies and Noninvasive Disease Biomarkers CHAPTER 490
■ NIPT FOR FETAL MICRODELETION/
DUPLICATION SYNDROMES
Advanced molecular technologies, particularly chromosomal microarray (CMA), are now the preferred diagnostic method when performing amniocentesis or CVS, particularly in the setting of a known fetal
anomaly or fetal growth restriction. Unlike traditional karyotype,
which relies on the visual assessment of chromosomes by a cytopathologist with a resolution of ~5–10 MB, CMA is an automated molecular
technique that can identify differences as small as a single nucleotide.
The main benefit of CMA over traditional karyotype is in its ability to
detect copy number variants such as microdeletion/duplication syndromes (e.g., 22q11 microdeletion or DiGeorge’s syndrome). Although
the focus of NIPT was initially on the prenatal detection of Down’s syndrome and other aneuploidies, the potential for NIPT to detect smaller
chromosomal rearrangements, including copy number variations
(CNVs), was recently evaluated and introduced into clinical care. This
is important because CNVs carry a significant neonatal disease burden, and the risk of such variants may be more common than Down’s
syndrome among young pregnant women. Unfortunately, the detection rates for such conditions by NIPT are not as robust as they are
for aneuploidy detection, and the positive predictive values are lower.
Moreover, because most cell-free fetal DNA originates from the placenta, there is concern that confined placental mosaicism may result in
erroneous NIPT results that target smaller genomic aberrations. High
false-positive rates may lead to unnecessary parental anxiety, increased
rates of invasive confirmatory testing, and pregnancy termination.
■ WHOLE GENOME NIPT AND
SINGLE-GENE DISORDERS
Most NIPT platforms use next-generation sequencing to obtain DNA
reads covering the entire genome but only report a fraction of the
sequences analyzed. This has led to a debate as to whether whole
genome NIPT should be offered and reported. As with any robust and
comprehensive test, questions regarding false-positive rates and abnormalities of unclear significance are being considered. Also, whether to
limit the results to known disease-causing mutations remains controversial. While whole genome NIPT is being considered, NIPT platforms targeting single-gene disorders have been recently developed,
and these are slowly becoming clinically available. This is a significant
advance because maternal carrier screening of single-gene disorders
is becoming more prevalent, with clinically available comprehensive
gene panels covering hundreds of potential disease loci. In the absence
of NIPT, couples deemed at risk based on maternal and/or paternal
carrier screens (e.g., an asymptomatic couple in which both parents are
heterozygous for an autosomal recessive gene mutation such as cystic
fibrosis) would need to undergo an invasive diagnostic procedure in
order to confirm or exclude a significant fetal condition (homozygous for
an abnormal cystic fibrosis gene).
■ NIPT USING CELL-FREE RNA
Cell-free fetal DNA is not the only nucleic acid found in maternal
serum. Cell-free fetal RNA is also present, providing a unique snapshot
of which genes are being transcribed at any given time in pregnancy.
Cell-free fetal RNA can also be linked to tissue-specific genes, offering
a unique window into developing fetal organs. Moreover, serial analysis
of the pregnancy transcriptome and comprehensive analysis of cfRNA
in maternal serum over time have been studied as means of early
detection of common third-trimester obstetric complications including
preeclampsia, fetal growth restriction, and preterm birth.
■ NIPT DETECTING MATERNAL MALIGNANCY
Maternal malignancy complicates approximately 1 in 1000 to 1 in
1500 pregnancies. Common cancers include hematologic, breast,
and gynecologic cancers, including cancers of the cervix and ovary.
Because next-generation technologies sequence both maternal and
fetal DNA, changes in DNA levels may not only indicate fetal disorders
but also maternal disease. While most uninterpretable NIPT tests result
from low fetal fraction or other fetal and/or placental conditions, a
small subset may be due to undiagnosed maternal malignancy. Cases
of maternal lymphoma, leukemia, gastrointestinal malignancy, and
neuroendocrine tumors detected incidentally by NIPT have been
described in the literature.
■ BEYOND NEXT-GENERATION SEQUENCING
While most NIPT platforms use next-generation sequencing technology, some studies have analyzed the utility of digital PCR and fetal
cell analysis for NIPT. Digital PCR has been described as a useful tool
for testing pregnancies at risk of X-linked and autosomal recessive
single-gene disorders for both single mutations and compound heterozygous mutations. Although original attempts to isolate circulating fetal
cells were abandoned >20 years ago, recent advances have led to new
technologies targeting both circulating fetal nucleated red blood cells
and trophoblasts. This progress, still in the research phase, suggests that it
may be possible to identify and isolate single cells as early as the first trimester and analyze them by whole-gene genome amplification followed
by copy number analysis using arrays or next-generation sequencing.
SUMMARY
Collectively, the uses described in this chapter summarize some of
the myriad clinical applications of liquid biopsy methods including
cfDNA profiling in oncology, transplantation medicine, and obstetrics.
For each area, several existing assays have enabled transformative
approaches to noninvasive diagnosis of disease, assessment of associated risk, and assessment of therapeutic responses. For example,
several tumor-derived cfDNA assays are currently available for early
cancer detection, noninvasive genotyping, response assessment, and
MRD monitoring across diverse cancer types. Liquid biopsy assays for
oncology can provide highly sensitive measures of tumor burden and
associated risk and can also be used to monitor response to anticancer
therapies. Prominent among applications of liquid biopsies is the detection of MRD following definitive therapy for solid cancers as a strongly
prognostic biomarker with very high positive predictive value for
risk of recurrence. Similarly, cell-free nucleic acids have transformed
transplantation medicine. Rising levels of donor-derived cfDNA can
indicate acute allograft rejection, and clinical dd-cfDNA assays are
now being used for noninvasive rejection surveillance in lieu of invasive biopsy procedures. Microbial cfDNA analysis can potentially be
used to screen for opportunistic infections after transplantation and
for the hypothesis-free diagnosis of posttransplant infections, and
such clinical assays are currently in development. Moreover, quantification of particular microbial cfDNA sequences, such as that of the
Anelloviridae family, could one day enable transplant physicians to
assess the overall status of the alloimmune response—a goal that could
enable personalization of immunosuppressive therapy to maximize
efficacy and to prevent the long-term toxicities associated with these
medications. Finally, detection of cell-free fetal or placental DNA has
improved the prenatal detection of fetal aneuploidy and other genomic
abnormalities. Accordingly, the potential of these liquid biopsy techniques is rapidly expanding and places us at the cusp of a revolution in
personalized management of patients across medical disciplines using
innovative noninvasive techniques.
■ FURTHER READING
Alix-Panabières C, Pantel K: Clinical applications of circulating
tumor cells and circulating tumor DNA as liquid biopsy. Cancer
Discov 6:479, 2016.
Bianchi DW, Chiu RWK: Sequencing of circulating cell-free DNA
during pregnancy. N Engl J Med 379:464, 2018.
Chabon JJ et al: Integrating genomic features for non-invasive early
lung cancer detection. Nature 580:245, 2020.
Crowley E et al: Liquid biopsy: Monitoring cancer-genetics in the
blood. Nat Rev Clin Oncol 10:472, 2013.
De Vlaminck I et al: Temporal response of the human virome to
immunosuppression and antiviral therapy. Cell 155:1178, 2013.
Khush KK: Clinical utility of donor-derived cell-free DNA testing in
cardiac transplantation. J Heart Lung Transplant 40:397, 2021.
Kurtz DM et al: Circulating tumor DNA measurements as early
outcome predictors in diffuse large B-cell lymphoma. J Clin Oncol
36:2845, 2018.
3846 PART 20 Frontiers
Kurtz DM et al: Dynamic risk profiling using serial tumor biomarkers
for personalized outcome prediction. Cell 178:699, 2019.
Moding EJ et al: Detecting liquid remnants of solid tumors: circulating
tumor DNA minimal residual disease. Cancer Discov 2021.
Morain S et al: A new era in noninvasive prenatal testing. N Engl J
Med 369:499, 2013.
Warsof SL et al: Overview of the impact of noninvasive prenatal testing on diagnostic procedures. Prenat Diagn 35:972, 2015.
Many hundreds of human diseases, collectively known as protein conformational diseases or protein folding disorders, result from protein
misfolding due to intrinsic and extrinsic errors amplified by exposures
to environmental and physiologic stress conditions. Despite many
years of considerable effort, there remains no useful algorithm that can
effectively predict protein tertiary (three-dimensional) structure from
primary amino acid sequence (and its variants), let alone posttranslationally modified (from environmental exposures) primary sequence.
Such events challenge the integrity of the proteome and can lead to
premature clearance, mislocalization, dysfunction or aggregation of
proteins, thus affecting cellular robustness, health, and longevity. Mismanagement of the proteome is the basis of a broad class of hundreds
of diseases that include orphan lysosomal storage diseases, type 2
diabetes, cystic fibrosis, certain fibrotic diseases, metabolic diseases,
muscle wasting diseases, cancer, and neurodegenerative diseases, as
exemplified by Alzheimer’s disease, frontotemporal dementia, Parkinson’s disease, amyotrophic lateral sclerosis (ALS), and Huntington’s
491 Protein Folding Disorders
Richard I. Morimoto, G. Scott Budinger
Eye Neuronal tissue
Lung
Pulmonary alveolar
proteinosis
Cystic fibrosis
Thyroid
Medullary thyroid carcinoma
Immune system
Systemic AL amyloidosis
Multiple myeloma
Cataracts
Corneal lactoferrin
amyloidosis
Hereditary lattice
corneal dystrophy
Alzheimer’s disease
Amyotrophic lateral sclerosis
Familial British dementia
Familial Danish dementia
Parkinson’s diease
Huntington’s disease
Muscle
Inclusion body
myositis/myopathy
Aortic medial
amyloidosis
Cardiac amyloidosis
(e.g., transthyretin
cardiomyopathy)
Liver
Systemic diseases
Type 2 diabetes mellitus
α1 Antitrypsin deficiency
Lysozyme storage diseases
p53-dependent cancers
Pancreas/islet a cells
FIGURE 491-1 Diseases of protein folding. A representative list of tissues and known folding diseases.
disease (Fig. 491-1). For each of these diseases and many others
described in this textbook, aging is the major contributing risk factor.
The challenge at the biochemical and molecular level is for the cell
to achieve a stable and functional proteome during development that
persists through young adulthood and to maintain it throughout aging.
For humans, this is necessary for the operational health of each of the
tens of trillions of cells that compose our ~80 organs for health span
and life span. To achieve this goal, our cells have evolved a remarkably
efficient proteostasis network (PN) composed of molecular chaperones
and other highly conserved components essential for normal protein
synthesis, folding, translocation, and degradation (Fig. 491-2) that
balances input with output and that ensures every protein is functional.
The PN is essential for all tissues and for the diverse protein-protein
interactions required for cell signaling, biosynthetic processes, and
the structural demands for tissue shape, mechanical properties, and
function. An equal, if not more important, role for the PN is to detect,
prevent, and remove misfolded and aggregated proteins that accumulate in stress, aging, and disease and that thereby interfere with cellular
health. Understanding how proteostasis is achieved and maintained is
of fundamental biological interest and essential to prevent age-associated
protein folding disorders.
All organisms use an evolutionarily conserved set of molecular
machines for the synthesis, folding, transport, and removal of unnecessary and damaged proteins. The PN evolutionarily has adapted to the
highly specific physiologic requirements of tissues and the expression
of abundant and rare proteins with wide-ranging solubilities, folding
requirements, stabilities, and structural demands. Added to this complexity of natural clients for the PN is the additional load generated by
genetic mutations carried in natural populations and environmental
stressors that challenge the capacity of the PN. Despite the central role
of proteins as the workhorse of the cell, they are highly susceptible to
molecular damage, whether due to intrinsic metastability or genetically
inherited mutation or because of error-prone synthesis. Hence, dysfunction in the PN may clinically manifest as either a gradual decline
in homeostatic function as occurs with genetic mutations or a loss of
resilience in the face of environmental stressors. Thus, clinicians see the
consequences of proteostasis failure and cellular dysfunction in both the
myriad disorders that present to them as
age-associated clinical problems and the
increased morbidity and mortality associated with trauma, infection, and other
acute illness requiring hospitalization in
older individuals.
PROTEIN QUALITY
CONTROL MECHANISMS
The PN monitors and controls the flux
of protein synthesis in order to promote functional folding and to minimize the accumulation of off-pathway
aggregation-prone intermediates by
their selective disaggregation or degradation. However, unlike an automobile
assembly plant for which each item is
designed and engineered for assembly
into functional components, the properties of the PN tolerate the tremendous
chemical noise and diversity generated
by coding region polymorphisms and
biosynthetic errors among its client
components, while maintaining the
ability to recognize and remove kinetically unstable conformational states of
proteins that would compromise assembly and function. Proteins are highly
sensitive to fluctuations in their intracellular environment caused by shifts in
energetics, pH, oxidizing and reducing
conditions, and the myriad of small
3847Protein Folding Disorders CHAPTER 491
Consistent with the recognition that the formation of off-pathway
aggregates is a kinetic component of proteostasis are the concerted
activities of disaggregase machines that can unravel protein aggregates
into the unfolded, extended polypeptide chain. These disaggregases
correspond to the AAA+ proteins, corresponding to ClpB in bacteria
and Hsp104 in yeast and plants and functionally related to a redirected
HSP70 machine in other eukaryotes and metazoans that interacts with
HSP110 and specific combinations of J-domain proteins capable of
inducing disaggregation.
The subcellular organelles, the mitochondria, and endoplasmic
reticulum (ER) are responsible for ~20% of the proteome. Chaperone
interactions are essential to maintain the extended polypeptide chain
in a recognition-competent state for the respective organellar receptors required for translocation across membranes. Upon import, each
translocated polypeptide is met by organellar-specific chaperones of the
HSP70 and J-domain family for folding and assembly. While the mitochondrial genome encodes 13 proteins required for electron transport,
the great majority of mitochondrial proteins are encoded by the nuclear
genome, synthesized in the cytosol, and then imported across the outer
and inner mitochondrial membranes. Hence, maintenance of the mitochondrial proteome relies on both the cytosolic and mitochondrial PN.
For translocation into the lumen of the ER, the extended polypeptide
interacts with a set of glycosyltransferases, calnexins, calreticulins and
disulphide isomerases. Proteins that misfold in the ER are recognized
and retrotranslocated to the cytoplasm, where they are directed to the
ubiquitin-proteasome system (UPS) for unfolding and degradation.
The PN is balanced by the essential catabolic processes of the
ubiquitin-proteasome and the autophagy-lysosomal machines that
recognize proteins for degradation and recycling. The UPS is generally
considered the primary pathway by which most proteins are recognized
and tagged for degradation; the autophagy-lysosome system is highly
responsive to nutrient changes and damage, recognizing and engulfing
organelles and other subcellular compartments and large aggregates
and inclusions. In addition to their role in the regulated turnover of
cellular proteins, these degradation systems are essential for protein
quality control and for limiting the accumulation of misfolded and
aggregated proteins during stress conditions, aging, and disease.
Protein turnover by the UPS involves an enzymatic cascade of E1,
E2, and E3 enzymes that utilize ubiquitin to tag clients, followed by
degradation of the polyubiquitinated substrates by the 26S proteasome.
Client specificity involves the large family of ~600 ubiquitin ligases.
In addition to their roles to mark proteins for degradation, the ubiquitination machinery has numerous additional functions in cellular
processes. For example, the ubiquitin ligase listerin is associated with
the ribosome and ubiquitinates nascent chains that stall in translation
to prevent the accumulation of aberrant polypeptides that would subsequently aggregate. Ubiquitination of these nonnative clients by the
ubiquitin ligase activity of the co-chaperone CHIP is central to the triage decision of the HSP70/HSP90 complex between client folding and
proteasome-mediated degradation. ER-targeted clients that are misfolded are retrotranslocated to the cytoplasm, polyubiquitinated, and
degraded by cytosolic proteasomes in a process termed ER-associated
degradation (ERAD). Ubiquitination also provides cross-talk between
the proteasome and autophagy pathways by targeting clients for lysosomal degradation and for endosomal sorting. Chaperones co-label
a protein as nonnative, recruiting other proteins that place ubiquitin
chains on the damaged protein for degradation by the proteasome.
Alternatively, chaperones can label proteins or protein aggregates to
target them to the lysosome. In this process, damaged proteins are
degraded by the lysosome, an intracellular organelle with an acidic
environment enriched in proteases, through autophagy.
While there is a good general understanding of the process of in vivo
chaperone-dependent protein folding, the details of how decisions are
made for each client in the cell—whether and for how long to be maintained in a nonnative folding state through chaperone interactions in
a nucleotide-independent state, or to assemble into a stable chaperone
complex for subsequent assembly into a functional state, or to interact
with chaperones to fold directly to a native state—remain to be fully
addressed.
molecules and metabolites that affect folding and function. Added to
this range of perturbations are the effects of external stress caused by
elevated temperatures, infections, abnormal redox environments, or
osmolytes that can have profound consequences on protein-folding
thermodynamics, kinetics, and function. These intracellular and extracellular stress conditions, if not properly addressed, would be predicted
to amplify further protein instability from sequence polymorphisms
and biosynthetic errors that contribute to the stress of protein misfolding. The PN is organized at the cellular level into a series of highly
coordinated molecular machines that direct the expression, biogenesis,
and functional health of essentially all proteins (Fig. 491-2). Serving
more than as regulator and orchestrator of these highly synchronized
events, the PN is essential for protein quality control and prevention
of the appearance of off-pathway conformational states and accumulation of aggregates and amyloid species. Proteome health involves the
constant exchange between the intrinsic physicochemical properties of
polypeptides and the biological milieu of the cellular environment in
which protein sequences and function evolved.
Beginning with the synthesis of the nascent polypeptide on the
ribosome, ribosome quality control (RQC) factors and cytoplasmic
chaperones of the HSP70, HSP90, DNAJ/HSP40, chaperonin/HSP60,
and small heat shock proteins (sHSPs) family ensure co-translational
and posttranslational folding for the cell. Approximately 60% of the
proteome resides in the cytoplasm and nucleus, for which the RQC,
HSP70, HSP90, and HSP60 chaperones regulate the folded state of
client proteins, together with co-chaperones, through cycles of ATP
binding and hydrolysis. Chaperones of the HSP70 and J-domain family
are particularly well studied for their ability to interact transiently with
nascent polypeptides in protein synthesis through short, dispersed,
hydrophobic regions using the energy from nucleotide hydrolysis to
stimulate the release of partially folded intermediates that either reenter the chaperone cycle or are in a folded native state. For other clients,
such as transcription factors, kinases, phosphatases, and signaling
molecules, folding to the functional state is regulated and dependent
upon interactions with the HSP90 chaperone and other regulatory
co-chaperones to form stable heteromeric complexes to maintain
the client in a partially folded state primed for subsequent regulated
release.
Intermediate Native state
folded states
Misfolded states
Molecular
chaperones
Chaperones
Autophagy
Proteasome
Unfolded nascent
polypeptide
Normal turnover
Improper trafficking Toxic folds Degradation
Emphysema Amyloidoses Cystic Fibrosis
Cystic fibrosis
transmembrane
conductance regulator
Abeta, tau, Huntington,
SOD1, α-synuclein
α1 Antitrypsin
FIGURE 491-2 The proteostasis network and protein folding diseases. The process
of protein biogenesis involves the action of molecular chaperones to ensure the
transition of the unfolded nascent polypeptide through intermediates to the folded
native state. Such proteins then have a natural turnover. Off-pathway species are
prevented by the actions of chaperones and the recognition of nonnative misfolded
states and aggregates by the autophagic-lysosomal machinery and the ubiquitinproteasome. When misfolded species escape quality control, they can then become
improperly trafficked, as occurs for α1
antitrypsin associated with emphysema; for
toxic folds as occurs for amyloid beta, tau, huntington, and SOD1 in amyloidogenic
neurodegenerative diseases; or prematurely degraded as occurs for cystic fibrosis
transmembrane conductance regulator (CFTR) associated with cystic fibrosis.
3848 PART 20 Frontiers
CELL STRESS RESPONSES: SENSORS AND
REGULATORS OF PROTEIN DAMAGE
Cell stress responses are ancient genetic networks that detect, adapt to,
and protect all cells against toxic environmental stressors and physiologically relevant changes in their cellular environment (Fig. 491-3).
At the core of these cell stress responses are molecular switches: (1) the
heat shock response (HSR) that protects proteins in the cytoplasm and
nucleus controlled by HSF-1; (2) the unfolded protein response (UPR)
of the ER (UPRER) controlled by XBP1, ATF6, and ATF4; (3) the UPR
of the mitochondria (UPRmito) controlled by ATFS1; (4) the DAF-16/
FOX-O stress response pathway associated with insulin signaling; and
(5) the antioxidant stress response regulated by NRF-2. Collectively,
these cell stress responses and their respective transcription factors are
each essential for all cells and tissues and are regulated both autonomously and cell nonautonomously across tissues in metazoans to detect
proteotoxic stress and to adapt to and protect the cell against the toxic
consequences of protein damage. While each of these cell stress pathways can be activated independently, they are also induced in different
combinations according to the chemical and physiologic properties of
the stress signal(s) and provide cross-protective mechanisms.
The HSR is an evolutionarily conserved cellular defense mechanism
that protects cells against proteotoxicity associated with misfolding,
aggregation, and proteome mismanagement. HSF-1 inducibly regulates
transcription of genes encoding molecular chaperones and components of the PN. In unstressed cells, HSF-1 is cytoplasmic or nuclear
and exists in an inert monomeric state negatively regulated by weak
interactions with the molecular chaperones HSP70 and HJD-1. Upon
heat shock stress, HSF-1 rapidly trimerizes to acquire DNA-binding
activity and undergoes extensive posttranslational modifications, binds
to heat shock elements in promoters of stress responsive genes, and
relocalizes into nuclear stress bodies. Upon dissipation of the stress
signal, attenuation of the HSR involves the active repression of HSF-1
DNA binding through acetylation and loss of HSF-1 transcriptional
activity by reassociation with HSP70 and other molecular chaperones
and with HSBP1, leading to dissociation to the monomeric inert state.
In addition to HSF-1 being essential for the HSR and cell and organismal stress resilience, HSF-1 is essential during early development in
metazoans, functions as a maternal factor for gametogenesis, regulates
oocyte maturation by activating genes that function in the meiotic cell
cycle, is constitutively activated in cancer, and is necessary to maintain
NAD+ and ATP levels.
In the ER, the UPRER involves three stress arms regulated by the TFs,
XBP1, ATF6, and ATF4, that bind to specific cis-elements for these
ER-stress-responsive arms. XBP1 is activated by the transmembrane
endoribonuclease IRE1, which is a transmembrane protein with kinase
and endoribonuclease (RNase) activity that senses misfolding in the
ER directly, leading to autophosphorylation, oligomerization, and
acquisition of RNase activity. These events allow active IRE1 to cleave
XBP1 mRNA, generating a spliced transcript (XBP1s) that encodes
XBP1, which induces the transcription of UPR target genes. ER stress
also promotes the relocalization of ATF6 from the ER membrane to the
Golgi apparatus, where it is cleaved by the proteases SP1 and SP2, generating a cytosolic fragment of ATF6 that translocates to the nucleus
to direct transcription of a complementary set of UPR genes. Together,
XBP1 and ATF6 induce the expression of genes involved in protein
folding, ER-associated protein degradation, and lipid metabolism
pathways. A third ER transmembrane protein, PERK, also induced by
ER stress, promotes translation of the TF ATF4 by phosphorylating
the translation initiation factor eIF2α. Under these conditions, ATF4
mRNA is preferentially translated, leading to selective expression of
the proapoptotic TF CHOP, which elicits apoptosis in cells in which
ER stress is not resolved, presumably to remove damaged cells from
the population.
For mitochondria, the UPRmito response involves ATFS1, which
contains a mitochondrial targeting sequence and a nuclear localization signal. Under normal cellular conditions, ATFS1 is imported into
mitochondria and degraded, but upon mitochondrial stress, ATFS1 is
directed only to the nucleus to regulate transcription of genes encoding mitochondrial chaperones, mitochondrial import machinery, and
glycolysis. In mammals, the UPRmito is regulated by ATF5, which corresponds to ATFS-1 in Caenorhabditis elegans.
In metazoans, the integration of stress survival strategies includes
the antioxidant factor SKN-1/NRF2, the insulin-signaling factor DAF16/FOXO, and the tissue identity factor PHA-4/FOXA. Oxidative
and xenobiotic stresses activate OxR, which controls the expression
of redox-regulatory proteins and components of protein degradative pathways mediated in mammals by NRF1/NFE2L1 and NRF2/
NFE2L2, which corresponds to SKN-1 in C. elegans. NRF1 is an
ER-resident factor that undergoes regulated proteolytic cleavage upon
activation to control expression of genes encoding subunits of the proteasome and the UPS. NRF2 in the cytoplasm is negatively regulated by
the redox-sensitive ubiquitin ligase KEAP-1; consequently, inactivation
of KEAP-1 by oxidative and electrophilic stress leads to stabilization
and nuclear translocation of NRF2, which, in turn, induces the expression of antioxidant proteins and detoxification enzymes.
ORGANISMAL PROTEOSTASIS IN
AGING AND DISEASE
Much of our understanding on protein quality control mechanisms
has come from in vitro studies with purified molecular chaperones
or components of the UPS, complemented with cell extracts and cellbased assays using yeast or mammalian cells grown in tissue culture.
A common theme that emerges from these studies is that of hormesis,
in which transient bouts of activation of the HSR, UPRER, or UPR mito
improves lifespan and organismal resilience but chronic activation is
detrimental.
The importance of these pathways is further highlighted by studies
suggesting neuronal coordination of stress responses at the level of the
organism. When neuronal mechanisms fail, the HSR reverts to cell-autonomous control. At the organismal level, however, the HSR, UPRER,
and the UPR mito in C. elegans are regulated by cell-nonautonomous
control through neuronal signaling. When neuronal signaling is
impaired, the HSR reverts to cell-autonomous control. Neuronal signaling also regulates the UPR mito with disruption of mitochondrial
function in C. elegans neurons activating the UPR mito in nonneuronal
tissues, supporting a role for a mitokine signal. Perturbation of the
mitochondrial electron transfer chain (ETC) was shown to increase
life span in both invertebrates and rodents through the activation of
the UPR mito. The response to mitochondrial dysfunction in C. elegans depends upon the severity of mitochondrial impairment with a
mild reduction of ETC having hormetic effects on organismal stress
resilience, proteostasis, and longevity by resetting the cytoplasmic
High
Risk
Development Reproduction Aging Disease
Programmed
Repression of
the Heat Shock
Response, UPR,
and Oxidative
Stress Response
Stress responses
Molecular chaperones
Protein quality control
Proteostasis
Low
Risk
FIGURE 491-3 Aging and protein folding diseases. Aging is the major risk factor
for degenerative diseases. Cell stress responses (heat shock response and the
unfolded protein responses in the endoplasmic reticulum and mitochondria) decline
at reproductive maturity in studies from Caenorhabditis elegans and prevent
adaptive and protective increased expression of molecular chaperones to prevent
protein misfolding. UPR, unfolded protein response.
3849Protein Folding Disorders CHAPTER 491
HSR through HSF-1, independent of ATFS-1 and the UPR mito.
Mild perturbation of the ETC in Drosophila muscle also has systemic
benefits on organismal health and life span involving the insulin signaling. Communication between neurons also regulates the UPRER
in peripheral tissues of C. elegans. During infection of C. elegans by
pathogens, induction of the UPRER in nonneuronal tissues is mediated
by sensory neurons, suggesting an organismal stress response. Cellnonautonomous regulation of the UPRER has also been observed in
mice, where overexpression of active XBP1 in pro-opiomelanocortin
neurons activates the UPRER in the liver.
Other forms of intertissue communication that regulate proteostasis
with beneficial effects on organismal health include: cholinergic signaling across the synaptic junction to enhance or inhibit HSF-1-regulated
proteostasis in the muscle cells of C. elegans; transcellular chaperone
signaling between somatic tissues or between somatic tissues and
neurons of C. elegans to regulate the expression of HSP90 in receiving
tissues via the tissue code factor PHA4/FOXA, protection of neurons
and glial cells from elevated temperature-induced death by overexpression of small HSPs in Drosophila flight motor muscle cells, enhancing
proteostasis in C. elegans muscle cells by elevated expression of DAF16/
FOXO in the intestine, and that overexpression of dFOXO/4E-BP
in Drosophila muscle influences proteostasis in retina, brain, and
adipose tissues to delay the age-dependent accumulation of protein
aggregates.
Cell stress responses and proteostasis decline in aging. Insights
on the relationship between proteostasis, cell stress, and aging have
come primarily from C. elegans with support from other invertebrate
and vertebrate model systems and human cells. A set of endogenous
metastable proteins that exhibit temperature-sensitive properties were
shown to misfold in C. elegans at the permissive temperature during
aging, which was associated with a decline in the HSR. The decline of
proteostasis in C. elegans aging is regulated via cell-nonautonomous
control involving germline stem cells that initiate the programmed
repression of the organismal HSR resulting in the loss of stress resilience and age-associated protein aggregation. This is regulated at
reproductive maturity by an epigenetic switch involving the timed
placement of repressive H3K27me3 chromatin marks at the heat shock
genes, causing chromatin inaccessibility for HSF-1. This age-dependent
decline in the HSR can be reversed either by blocking the signal from
germline stem cell signal(s) or preventing the epigenetic repressive
marks. The relationship between reproduction and inducibility of
the HSR observed in animals at reproductive maturity suggests that
the age-associated events of cellular failure and loss of tissue robustness
during aging are not random processes but, rather, highly regulated,
perhaps to ensure that somatic tissues are programmed to decline after
reproduction, consistent with the germline soma theory of aging.
Proteostasis is one of the pillars of gerontologic biology, which,
together with genomic instability, telomere attrition, epigenetic alterations, deregulated nutrient sensing, mitochondrial dysfunction, cellular
senescence, stem cell exhaustion, and altered intercellular communication, provides a mechanistic basis for the biology of aging. The programmed decline of proteostasis in early adulthood would suggest that
failure in protein quality control would have negative consequences on
the other key elements of gerontologic biology. Whether proteostasis
collapse is the first to fail or among the earliest events to fail in aging,
it is consistent with the very large number of human degenerative diseases in aging associated with protein misfolding.
PROPERTIES OF PROTEIN
FOLDING DISEASES
The complexity that arises with protein folding diseases is that all
tissues are at risk and essentially all proteins are at risk for misfolding.
Added to this is the effect of aging and that each protein folding disease
exhibits a highly variable age of onset for pathology. There is additional
complexity in classification—whether to organize folding diseases by
tissues, i.e., muscle proteinopathies or neurodegenerative diseases,
according to the specific protein that misfolds such as α1
antitrypsin
(AAT) deficiency, or by the biophysical nature of the aggregate species
in systemic amyloidoses.
Disorders in which a specific mutation leads to protein misfolding or
the formation of a specific insoluble protein aggregates likely represent
only the tip of the iceberg of protein folding disorders. Mutations in
aggregation-prone proteins coupled with changes in the cellular environment and decline in capacity and robustness of the PN in aging
promote protein misfolding and aggregation in affected tissues. Once
aggregation has initiated, this leads to further impairment in protein
quality control pathways, causing further collateral damage and aggregation of other at-risk proteins. Such a mechanism may only manifest
clinically after a seemingly random systemic stress like pneumonia,
large bone fractures, or ischemic vascular events, possibly explaining
the rapid (1–2 years) accumulation of age-related morbidities in the
year following the first event. As such, the age-related decline in the
function of any of the components of the PN could underlie the compounding multiple morbidity that limits health span and life span in
many elderly individuals. Within this framework, it is useful to discuss
some of the better understood mechanisms of proteostasis dysfunction
that have been causally linked to diseases in humans.
■ DISORDERS THAT ENHANCE MISFOLDING AND
CAUSE PREMATURE DEGRADATION
(CYSTIC FIBROSIS)
Cystic fibrosis (CF) is a recessive disorder caused by mutations in both
alleles of the cystic fibrosis transmembrane conductance regulator
(CFTR) gene that encodes a multidomain membrane-spanning chloride ion channel protein. Thousands of mutations in CFTR have been
identified that affect CFTR biosynthesis, folding, trafficking, and function, leading to chronic obstructive lung disease, intestinal obstruction,
liver dysfunction, exocrine and endocrine pancreatic dysfunction, and
male infertility. CF is a folding disease due to its recognition by the
PN as misfolded protein. The most prominent mutation is deletion of
phenylalanine 508 (F508del), present in ~90% of CF patients. Mutant
ΔF508 retains partial channel function, but because it is recognized as
misfolded in the ER and the cytoplasm, it is marked with ubiquitin for
degradation by the ubiquitin proteasome system. Small molecules that
affect the conformation and function of mutant ΔF508 channel function can result in substantially improved outcomes in patients.
■ DISORDERS THAT INDUCE TOXIC AGGREGATES
AND LOSS OF FUNCTION (AAT DEFICIENCY)
AAT deficiency (AATD) is a co-dominant inherited disease with an
increased risk of chronic obstructive pulmonary disease (COPD), liver
disease, and vascular inflammation. Pulmonary problems are more
frequent in adults, whereas liver and skin problems may occur in adults
and children. AAT is encoded by the SERPINA1 gene, secreted into the
circulation by the liver, and responsible for inactivating endogenous
proteases, particularly those secreted by neutrophils and other inflammatory cells in the lung. Patients with AATD harbor mutations in
SERPINA1 that cause misfolding in the ER. The two major phenotypes
resulting from this abnormality highlight the diverse consequences
of misfolding on different cells and organs. In the liver, misfolding of
the mutant protein results in the formation of toxic aggregates and
hepatocyte death, manifest as liver injury and eventually cirrhosis—a
gain-of-function toxicity. In the lung, the failure to secrete sufficient
AAT causes unchecked proteolytic damage to the delicate architecture
of the alveolus, a process that is markedly worsened when neutrophils
are recruited to the lung in response to cigarette smoking. This loss-offunction phenotype manifests pathologically as emphysema and clinically as COPD.
■ INTERACTIONS WITH PN COMPONENTS THAT
CHANGE CONFORMATION, STABILITY,
OR FUNCTION (CANCER)
Mutations in the tumor suppressor p53 are among the most common
mutations observed in patients with cancer. Deletion of p53 combined
with overexpression of an oncogene is sufficient to drive metastatic
No comments:
Post a Comment
اكتب تعليق حول الموضوع