Science Abm8108

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

C O VI D -19: 2 Y EA R S ON

REVIEW blood of SARS-CoV-2–infected individuals com-


pared with other respiratory viruses (10–12).
The immunology and immunopathology of COVID-19 IFN-III (IFN-l1 and IFN-l3) and ISGs are ex-
pressed in the upper airways of patients with
Miriam Merad1*, Catherine A. Blish2*, Federica Sallusto3,4*, Akiko Iwasaki5,6* reduced disease risk or severity, whereas expres-
sion of IFN-l2 and type I IFNs—but not ISGs—
Considerable research effort has been made worldwide to decipher the immune response triggered upon severe can be elevated in the upper airways of those who
acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, identify the drivers of severe and fatal develop severe COVID-19 (13). Patients with gene-
COVID-19, and understand what leads to the prolongation of symptoms after disease resolution. We review the tic mutations or autoantibodies that interfere
results of almost 2 years of COVID-19 immunology research and discuss definitive findings and remaining with IFN pathways suffer from life-threatening
questions regarding our understanding of COVID-19 pathophysiology. We discuss emerging understanding of COVID-19 disease as discussed below. During
differences in immune responses seen in those with and without Long Covid syndrome, also known as post-acute the late phase of disease, prolonged IFN sec-
sequelae of SARS-CoV-2. We hope that the knowledge gained from this COVID-19 research will be applied in retion correlates with worse disease outcomes
studies of inflammatory processes involved in critical and chronic illnesses, which remain a major unmet need. (14, 15), possibly through the induction of chemo-
kines that recruit inflammatory cellular infiltrates

I
(16). In addition, COVID-19 is accompanied by a
n December 2019, the Chinese Center for to spread more efficiently, making it difficult significant decline in the number of immune
Disease Control and Prevention reported a to distinguish increased infection rates from in- sentinel cells including pDCs and conventional
cluster of pneumonia cases with unknown creased severity. As new variants emerge, it will DCs (cDCs) in the blood (14) and lungs (17).
cause. The first complete sequence of the be important to direct continued research ef- Innate immune responses induced by PRR
novel beta coronavirus genome was then forts into identifying how such variants escape signaling activate effector cells to mediate viral
identified in January 2020 (1). Initially called from either innate or adaptive immune re- clearance. NK cells, for example, contribute to
2019-nCoV (for 2019 novel coronavirus), the sponses. Here, we review progress in our detecting and eliminating virally infected cells.
virus was renamed severe acute respiratory understanding of the immunology and immu- However, in patients with severe COVID-19, NK
syndrome coronavirus 2 (SARS-CoV-2) and nopathology of COVID-19. cells are depleted from the blood and become
the disease caused by the virus as coronavirus dysfunctional as a result of transforming growth
disease 2019 (COVID-19). COVID-19 manifests Innate immunity to SARS-CoV-2 factor–b (TGF-b), which impairs their antiviral
with symptoms ranging from fully asympto- Coronaviruses are enveloped viruses with function (18). Alveolar macrophages, which play
matic to severe disease and death (Table 1). a positive-sense RNA-strand genome of an important sentinel role in the lungs by sensing
The most common manifestations are fever, ~30,000 bases. SARS-CoV-2 is a member of and triggering potent antiviral immunity, are
cough, and shortness of breath; additional com- the Betacoronavirus genus, which also includes severely depleted in the lungs of patients with
mon symptoms include fatigue, myalgias, nau- severe acute respiratory syndrome coronavirus severe disease (19, 20). In contrast to the impaired

Downloaded from https://www.science.org on March 16, 2022


sea, vomiting, diarrhea, headache, weakness, (SARS-CoV) and Middle East respiratory syn- early antiviral defense mediated by IFN-I and
rhinorrhea, anosmia, and ageusia (2, 3). A wide drome coronavirus (MERS-CoV). The innate im- IFN-III, proinflammatory cytokines and chemo-
range of complications can lead to severe illness mune system uses various pattern recognition kines are significantly elevated (16). In addition,
and death, including pneumonia, acute respira- receptors (PRRs) to detect the presence of vi- T cells are profoundly depleted from the blood
tory distress syndrome, liver injury, cardiac in- ruses. RNA viruses such as coronaviruses are de- circulation of patients with severe disease com-
jury, thrombosis including stroke, renal disease, tected through two separate strategies (8). In one pared with patients with moderate disease,
neurologic disease, and sepsis (2–7). strategy, specialized immune cells such as plas- which suggests that induction of adaptive ef-
As the virus evolves and new variants emerge, macytoid dendritic cells (pDCs) detect the incom- fector T cells able to clear virally infected cells
there have been concerns that such variants ing viral genomic RNA in the endosome through is likely impaired in severe patients (14, 21–24).
could increase pathogenesis by escaping from Toll-like receptor 7 (TLR7). Other cell types ex- Pathological immune responses that contribute
immunity generated through previous infec- press endosomal TLR3 (variety of cells) and TLR8 to severe COVID-19 are discussed below.
tion or vaccination or by inducing more severe (myeloid cells) that can also recognize endocy-
disease. Some variants of concern (VOC), such tosed double-stranded RNA (dsRNA) or single- Adaptive immunity to SARS-CoV-2
as 1.351 (Beta), P.1 (Gamma), and the recently stranded RNA (ssRNA), respectively. Alveolar Given the key role of adaptive immunity in
described B.1.1.529 (Omicron), have muta- macrophages survey the lumen of the respira- protection from viral infection and disease,
tions that render them less susceptible to tory tract and provide the first line of defense. the study of humoral antibody (Ab) and cell-
vaccine-mediated and infection-acquired im- The other strategy involves viral recognition mediated immunity has been aggressively pur-
munity. It is less apparent whether some var- within infected cells. During viral replication sued by the scientific community with the aim
iants induce more severe disease upon primary dsRNA intermediates can be recognized by of defining correlates of protection, developing
infection than others, though strains such as cytosolic RNA sensors such as RIG-I and MDA5, immune-based therapies, and optimizing vac-
B.1.1.7 (Alpha) and B.1.617.2 (Delta) are known or RIG-I–like receptors (RLRs). Upon engage- cine design and administration (25–27).
ment of TLRs and RLRs, signaling downstream Abs can neutralize SARS-CoV-2 by blocking
1
Precision Immunology Institute, Tisch Cancer Institute, Icahn activates the IRF3/IRF7-dependent transcription binding of the viral spike (S) protein to the
School of Medicine at Mount Sinai, New York, NY 10029, USA.
2
Department of Medicine and Program in Immunology, Stanford
of type I and type III interferons (IFNs) as well ACE2 receptor and can promote effector func-
University School of Medicine, Stanford, CA 94305, USA. as nuclear factor kB (NF-kB)–dependent pro- tion by binding to the complement and Fc
3
4
Institute of Microbiology, ETH Zürich, 8093 Zürich, Switzerland. inflammatory cytokines and chemokines. receptors (28). Structure-guided serological
Institute for Research in Biomedicine, Università della Svizzera
SARS-CoV-2 is adept at evading innate rec- studies demonstrated that the bulk of neu-
Italiana, 6500 Bellinzona, Switzerland. 5Department of
Immunobiology, Yale University School of Medicine, New Haven, ognition, signaling, IFN induction, and IFN- tralizing Abs bind to distinct epitopes in the
CT, USA. 6Howard Hughes Medical Institute, Yale University stimulated genes (ISGs) through the expression receptor binding domain (RBD) of the S protein
School of Medicine, New Haven, CT, USA of a number of viral proteins that block these and primarily to sites 1a and 1b, which represent
*Corresponding author. Email: miriam.merad@mssm.edu
(M.M.); cblish@stanford.edu (C.A.B.); federica.sallusto@irb.usi.ch pathways (9). Consequently, lower levels of IFN-I the receptor-binding motif (RBM). Additionally,
(F.S.); akiko.iwasaki@yale.edu (A.I.) or IFN-III are detected in the lungs or peripheral a minor fraction of neutralizing Abs bind to the

Merad et al., Science 375, 1122–1127 (2022) 11 March 2022 1 of 6


N-terminal domain (NTD) (29–31). Further- for the postfusion conformation, which are eli- or abolished the efficacy of several approved
more, some RBM-specific Abs can mimic ACE2 cited by natural infection (47). Emerging SARS- mAbs—even when used in combination—
and directly trigger and consequently disable CoV-2 VOCs can, however, evade the neutralizing against the highly mutated Omicron variant
the fusion protein, a property that was initially Ab response elicited by infection or by current (51). The current strategy is to focus on Abs
discovered in SARS-CoV. Neutralizing epito- vaccines based on the Wuhan strain (Wuhan- that are less susceptible to variants because
pes outside RBD and NTD are generally sub- Hu-1) (48–50). Mutations found in VOCs primarily they mimic ACE2 or recognize conserved epi-
dominant but may be more conserved as in the cluster in the RBM, resulting in increased bind- topes such as S309/sotrovimab, which binds
case of the stem-helix region (32). The quantity ing to ACE2 and escape from neutralizing Abs. to RBD site IV outside of the RBM and retains
and quality of the Ab response is related to the Furthermore, mutations and deletions in NTD virus-neutralizing activity and effector func-
amount of antigen and the duration of the change the domain structure and may account tions against VOCs, including Omicron (51).
germinal center reaction where the response for differences in neutralizing titers observed
matures (33). Ab responses of increasing mag- against different VOCs (31). The Omicron variant Immunopathology of severe COVID-19
nitudes are detected in infected, preimmune, in particular carries 15 mutations in the RBD, SARS-CoV-2 shares 73% homology with SARS-
and vaccinated individuals, most likely reflect- which substantially erode the neutralizing activity CoV, and similar to SARS-CoV the pathogenesis
ing differences in antigenic load and exposure induced by previous infection or vaccination. of SARS-CoV-2-induced pneumonia likely occurs
(29, 34). In immunosuppressed patients and The interrogation of memory B cells has in two phases (52): First, the viral phase is charac-
those undergoing dialysis, Ab response is gen- provided a plethora of monoclonal Abs that terized by viral replication resulting in direct
erally poor and has been shown to contribute have been instrumental in dissecting the mech- virus-mediated tissue damage. The extent of this
to the chronic persistence of the virus favor- anisms of protection and informing vaccine damage determines the pathogenesis of the sec-
ing the selection of variants (35). design. Notably, several potent neutralizing ondary phase, which is characterized by the re-
CD4+ and CD8+ T cells elicited by SARS-CoV-2 mAbs have been rapidly developed and found cruitment of effector immune cells causing a local
infection are directed against a range of antigens to be effective as therapeutics when adminis- and systemic inflammatory response that can
including structural and nonstructural proteins tered to patients who may be at risk during early persist even after viral clearance. The development
and are significantly associated with milder dis- infection (27). One limitation of this approach of pulmonary disease is associated with excess
ease (36). Ab-mediated depletion of CD8+ T cells can be found in the variability of the S protein, vascular permeability leading to microthrombi
in convalescent macaques partially abrogates pro- particularly of the RBM, which has reduced deposition and vascular permeability (53), in
tection against rechallenge with SARS-CoV-2,
suggesting a role for CD8+ T cells in the face of
waning Ab responses (37). In view of its rele- Table 1. Common manifestations and potential complications of COVID-19. N/A, not applicable.
vance for the induction of neutralizing Abs, the
CD4+ T cell response to the S protein has been Acute and severe Covid Long Covid
studied in great detail in convalescent indi- Organ system Common symptoms Potential complications Common symptoms

Downloaded from https://www.science.org on March 16, 2022


viduals and vaccinees with prediction algo- Respiratory Cough Pneumonia Acute respiratory Dyspnea, cough, interstitial
rithms, peptide or protein stimulation, and Dyspnea, Rhinorrhea distress syndrome (ARDS) lung disease
isolation of T cell clones (38–40). These studies (runny nose) Respiratory failure
revealed multiple T cell epitopes in the RBD— Fever Sepsis, kidney injury Fever, fatigue
including an immunodominant site, which is Systemic
Fatigue Liver injury post-exertional malaise
highly conserved among Sarbecoviruses and
Nervous System Headache Stroke Memory deficit, loss of
VOCs. As discussed below, one of the limita- Confusion Seizure concentration, sleep difficulty,
tions imposed by severe COVID-19 is the pro- Anosmia Guillain-Barre mood disorder, paresthesia,
found T cell lymphopenia that affects the host’s (loss of smell) syndrome myelopathy, neuropathy,
ability to mount a robust immune response. Ageusia anosmia, ageusia, blurry
Abs and memory T cells elicited by common (loss of taste) vision, tinnitus, headache
cold coronaviruses are recalled by SARS-CoV-2 Cardiovascular Chest pain Myocardial infarction Chest pain, palpitations,
infection or vaccination, but their role in pro- Shortness of Myocarditis hypertension, angina,
tection or pathology remains to be established breath Cardiogenic shock myocarditis, pericarditis,
(41, 42). Further, it was suggested that preex- Stress-induced cardiac arrhythmias,
isting memory CD4+ T cells specific to an early cardiomyopathy postural orthostatic
transcribed replication transcription complex Clotting abnormalities syndrome
found in abortive infection may confer cross- Gastrointestinal Nausea Acute pancreatitis Diarrhea, vomiting,
protection against SARS-CoV-2 (43). In addi- Vomiting Acute appendicitis nausea, loss of
tion, cross-reactive Abs elicited by seasonal Diarrhea Intestinal obstruction appetite, hepatitis
coronaviruses may influence the course of the Anorexia (loss Bowel ischemia
disease as well as de novo immune responses of appetite) Hemoperitoneum
to SARS-CoV-2 (44, 45). Abdominal compartment
Seminal studies on RSV have paved the way syndrome
for the use of stabilized prefusion proteins as Skin Rash N/A Hair loss
vaccines to facilitate the production of neutral-
Musculoskeletal Myalgias N/A Myalgia, arthralgia,
izing Abs (46). Similarly, a prefusion stabilized S
(muscle aches) weakness
protein in the Moderna and Pfizer/BioNTech
mRNA vaccines and in the Ad26-based Johnson Endocrine Hyperglycemia Diabetes Hot/cold sensation, thyroid
& Johnson vaccine was used to optimize the abnormalities, diabetes,
production of neutralizing Abs while limiting hormonal abnormalities
the production of non-neutralizing Abs specific

Merad et al., Science 375, 1122–1127 (2022) 11 March 2022 2 of 6


C O VI D -19: 2 Y EA R S ON

Inability to mount a timely response Pathogenic inflammation

Plasmablast
Alveolar macrophage

SARS-CoV-2
Monocyte
T cell

B cell

Delayed IFN response Excess myeloid cell recruitment


Altered antigen presentation function Vascular damage, blood clots
Impaired alveolar macrophages Tissue damage

Increased with age, Virus


diabetes, obesity, persists
and other chronic Excess myelopoeisis and release of myeloid cells
inflammatory conditions
Vascular leakiness

Fig. 1. COVID-19 pathogenesis. Inability to mount a timely and effective site. Enhanced myelopoiesis and vascular damage common in older individuals and
antiviral response because of delayed IFN response, altered antigen presentation in patients with chronic inflammatory diseases further contribute to the enhanced
function, or altered tissue resident macrophage pool, common in older individuals, release of inflammatory myeloid cells from the bone marrow to the blood circulation
promotes viral persistence and prolonged tissue damage that trigger the prolonged and their recruitment to the site of infection leading to profound tissue damage,
blood release and recruitment of damaging inflammatory myeloid cells to the infected vascular lesions, and blood clots common in patients with severe disease.

addition to a range of systemic symptoms in- antiviral immunity. Loss of function variants less potent viral control while contributing to
cluding (i) olfactory dysfunction, which occurs in in loci that control TLR3- and IRF7-dependent tissue damage as suggested below.

Downloaded from https://www.science.org on March 16, 2022


approximately half of people diagnosed with type I IFN immunity have been identified in a
COVID-19 but has not been described for SARS- small number of severe patients (56). Notably, Inability to control SARS-CoV-2–driven
CoV (54); (ii) gastrointestinal (GI) symptoms these patients were adults who had never inflammatory responses
(55); and (iii) cardiac, hepatobiliary, and renal been hospitalized for severe viral illness before Autopsies of deceased COVID-19 patients have
dysfunction (53). The pathogenesis of extra- COVID-19. AutoAbs against IFN-a and IFN-w revealed very little active viral infection and
pulmonary manifestations is likely multifactorial (57–59) have been identified in patients with substantial accumulation of activated immune
as a result of direct viral injury to the tissue cells, severe disease and have been shown to con- cells, suggesting that organ failure is unlikely
vessels, or neurons, as well as cytokine release, tribute to delayed viral clearance (59). Neutral- to result from extensive viral-induced tissue
auto-Ab-induced tissue damage, vascular dam- izing IFN-a and IFN-w autoAbs have recently damage but is instead caused by an over-
age, or gut dysbiosis in the case of GI symptoms. been found to increase with age (60) suggesting activated immune system or vascular damage.
Although several risk factors for severe COVID- that type I IFN autoAbs can precede the disease Below, we discuss the potential causes of these
19 have been identified, the pathophysiological and serve as a biomarker of disease severity. excessive inflammatory responses.
underpinnings that contribute to disease sever- Imbalanced adaptive immunity: The reduc- Altered myeloid responses: Excess circulating
ity are not fully understood. Below, we discuss tion of DC numbers and profound T cell lym- immature monocytes, neutrophils, and mye-
two broad hypotheses that have emerged to phopenia driven by T cell sequestration in loid progenitors—referred to as emergency
explain severe COVID-19 pathophysiology and tissues or T cell apoptosis as a result of pro- myelopoiesis—are almost pathognomonic of
examine how the identified risk factors contrib- inflammatory cytokines is common in severe severe disease and are triggered during the first
ute to these pathophysiological drivers (Fig. 1). patients (22, 23) and may contribute to defec- phase of infection, likely because of delayed viral
tive T cell–mediated viral clearance. Defects clearance especially in conditions with preexist-
Inability to mount a timely antiviral response in type 1 immune responses (23) and excess ing altered myelopoiesis (62, 63). Circulating
The inability to mount a timely and effective type 2 immunity have both been shown to myeloid cells produce excessive amounts of in-
immune response against SARS-CoV-2 is multi- correlate with severe COVID-19 (14), suggest- flammatory molecules that promote vascular
factorial and is discussed as follows: ing that a maladapted adaptive immune re- permeability and organ damage (Fig. 1). By con-
Viral load: The exact contribution of viral sponse to the virus may also lead to delayed trast, lung tissue–resident macrophages known
ILLUSTRATION: V. ALTOUNIAN/SCIENCE

loads in disease course has been difficult to estab- viral clearance and disease progression. Sub- to play a key role in tissue homeostasis and repair
lish likely because of discrepancies in measure- stantial plasmablast expansion reaching up to are often depleted in severe patients (19, 20). The
ments between studies and sampling biases as 30% of circulating B cells—sometimes associated exact nature of their depletion is unclear, how-
well as viral dynamics, and it is still unclear how with extrafollicular responses—has also been ever, and may be a result of direct viral injury
much this contributes to disease outcome. reported in severe patients (61). The massive or cell death induced by excess inflammation.
Defects in type I IFN response: The timely plasmablast expansion may reflect polyreactiv- Pathogenic antibodies: AutoAbs have been
production of type I IFN by host cells is critical ity given the low levels of somatic mutations in identified in severe COVID-19 including a high
for limiting viral replication and promoting Ab clones observed in patients and may have proportion of Abs targeting nuclear antigen

Merad et al., Science 375, 1122–1127 (2022) 11 March 2022 3 of 6


(58, 59), phospholipids (64), T cell antigens, Immune-directed therapy for COVID-19 ongoing health problems that people experience
B cell antigens, chemokines, and cytokines Consistent with the major role of the immune 4 or more weeks after first being infected with
(57–59). Anti-phospholipid Abs cause clots in response in driving pathogenesis, a wide range SARS-CoV-2. By contrast, the World Health Or-
mouse models (64) whereas immunoglobulin of immunomodulatory agents have been tested ganization defines it as a condition that occurs in
G (IgG) from patients with anti-CD38 or anti- for the treatment of COVID-19, and several have people with confirmed or probable SARS-CoV-2
CD3e autoAbs exhibit increased Ab-dependent demonstrated efficacy. Evidence-based treat- infection, usually 3 months from the onset of
cellular phagocytosis by macrophages, which ment guidelines are regularly updated by the COVID-19 with symptoms and that last for at
may contribute to the profound lymphopenia National Institutes of Health and contain refer- least 2 months and cannot be explained by an
observed in some patients (59). AutoAbs have ences to all the primary literature guiding these alternative diagnosis (Fig. 2). A systematic review
also been detected in the cerebrospinal fluid recommendations (69). Such guidelines will con- of 57 peer-reviewed studies with 250,351 survi-
of patients with neurological symptoms (65). tinue to evolve as new drugs are developed vors of COVID-19 who met their inclusion crit-
In addition, afucosylated anti–SARS-CoV-2 and new clinical trials enable new indications. eria for PASC showed that the median age of
IgG1 with enhanced affinity for the activating The recent approval of the direct-acting anti- patients was 54.4 years, 56% were male, and 79%
FcgRIIIa accumulates mainly in male patients virals ritonavir-boosted nirmatrelvir (Paxlovid), were hospitalized during acute COVID-19 (70). At
with severe COVID-19 (66). These Abs fuel the remdesivir, and molnupiravir for nonhospital- 6 months, 54% of survivors suffered at least one
production of inflammatory cytokines and NK ized patients with mild-to-moderate COVID-19 PASC symptom. However, nonhospitalized
cell degranulation, which may contribute to who are at high risk of disease progression is an COVID-19 survivors who developed PASC were
tissue damage; furthermore, they are absent in excellent example of such a recent modification. primarily middle-aged women. In a survey of 445
asymptomatic patients and seropositive chil- Patients who are not hospitalized but are at high nonhospitalized Danish COVID-19 patients, per-
dren (66). The mechanisms that lead to the risk of disease progression can benefit from anti– sistent symptoms—most commonly fatigue and
production of afucosylated Abs and whether SARS-CoV-2 mAb products (either bamlanivimab difficulty with memory and concentration—were
these Abs can be induced upon vaccination plus etesevimab, or sasirivimab plus imdevimab reported by 36% of symptomatic participants with
still remains to be understood. or sotrovimab) (69). For hospitalized patients a follow up of >4 weeks. Risk factors for persist-
Vascular damage: Hypercoagulation, endo- who require supplemental oxygen, remdesivir ent symptoms included female sex (44% and
thelial damage, and arterial and venous embo- (a direct-acting antiviral) and dexamethasone 24% for women and men respectively, odds
lism are very common in severe COVID-19 (55). (a broad-spectrum corticosteroid) are recom- ratio 2.7) and Body Mass Index (odds ratio 1.1) (71).
Although the exact mechanisms of hyper- mended (69). The success of dexamethasone The immunobiology of PASC is currently un-
coagulability remain unclear, it may be a result in improving outcomes of patients with ad- der investigation. Leading hypotheses include
of direct viral damage to the vasculature or vanced disease highlights the critical role of (i) persistent virus or viral antigens and RNA in
severe inflammatory responses, which can inflammatory responses in mediating patho- tissues that drive chronic inflammation, (ii) the
alter the vascular endothelium and induce the genesis and marks a considerable departure triggering of autoimmunity after acute viral infec-
activation of platelets, monocytes, and macro- from the treatment of influenza, in which such tion (72), (iii) dysbiosis of microbiome or virome,

Downloaded from https://www.science.org on March 16, 2022


phages which in turn promote the release of steroid treatments are harmful. Indeed, several and (iv) unrepaired tissue damage (Fig. 2). SARS-
tissue factor, von Willebrand factor, and factor more targeted agents have also proven benefi- CoV-2 viral proteins and/or RNA have been de-
VIII, leading to the production of thrombin cial, particularly agents counteracting the IL-6 tected throughout respiratory, cardiac, renal, and
and fibrin clot formation (53). pathway including tociluzimab and sarilumab reproductive systems, as well as in the brain, mus-
(69). Inhibitors of the JAK–STAT signaling path- cles, eyes (73), GI tract (73–75), and lymph nodes
Predisposing risk factors way such as baricitinib and tofactinib have also (73, 75) months after infection. Aberrant innate
Age is by far the strongest risk factor for crit- shown promise (69). Drugs that target the IL-1 immune stimulation is associated with PASC
ical pneumonia with the risk of life-threatening pathway such as anakinra and canakinumab during the early and late phases of disease.
disease sharply increasing from 65 years of age have also been tested, though there is a lack of Studies have shown that some inflammatory
onward. Surprisingly, infants and young tod- sufficiently strong evidence of benefits to rec- cytokines—including IL-6, TNF-a, and IL-1b—
dlers commonly at risk for influenza infections ommend their use (69). Critically, IL-6- and JAK– are elevated in PASC patients (76, 77). IFN-b and
are usually protected from severe disease (67). STAT-blocking agents must be used within a brief IFN-l1 remain elevated 8 months after infection
Male patients are at much higher risk of de- window in recently hospitalized patients with in PASC patients compared with recovered
veloping severe COVID-19, which may be a re- rapidly declining courses. Presumably, this is controls. Furthermore, combinations of IFN-b,
sult of enhanced innate immunity and impaired when interruption of the “cytokine storm” can PTX3, IFN-g, IFN-l2/3, and IL-6 were associated
T cell activation compared with females (68) change the outcome of the disease. When such with PASC with 78.5 to 81.6% accuracy (78). A
and/or higher levels of afucosylated anti–SARS- anti-inflammatory drugs are used in less ill pop- deep, multiomic, longitudinal investigation of 309
CoV-2 Abs (66). Diabetes, hypertension, obesity, ulations or too late, there is a trend toward harm, COVID-19 patients (71% hospitalized) from initial
and chronic kidney diseases are also important making it imperative to identify those patients diagnosis to 2 to 3 months after identified four
risk factors for severe COVID-19. These condi- with inflammation that warrants intervention. PASC-anticipating risk factors at the time of in-
tions can include chronic inflammatory lesions, This opens the door for future diagnostics to bet- itial COVID-19 diagnosis: type 2 diabetes, SARS-
which may enhance and perpetuate the inflam- ter match a specific patient with a specific drug. CoV-2 RNAemia, Epstein–Barr virus viremia, and
matory cycle. Metabolic syndrome—often asso- autoAbs (79). AutoAbs were anticorrelated with
ciated with insulin resistance in diabetic and Immunology of Long Covid anti–SARS-CoV-2 Abs. These data collectively
obese patients—also leads to vascular damage It is now clear that COVID-19 can lead to long- suggest that the persistence of viral components
and impaired tissue repair. It is likely that the term disease—often referred to as Long Covid may result in chronically elevated IFNs and cy-
rapidity and quality of immune responses dur- syndrome or post-acute sequelae of SARS-CoV-2 tokines. Moreover, certain autoAbs may provide
ing the first phase of infection is particularly infection (PASC)—in a significant proportion of permissive conditions for such viral persistence.
critical in these patients as delayed viral clear- survivors. Although there is no universal consen- However, there is no causal link between viral
ance is more likely to lead to overt recruitment sus in the definition of PASC, the American proteins and RNA and these elevated cytokines.
and activation of immune cells, vascular dam- Centers for Disease Control and Prevention Adaptive immune responses to SARS-CoV-2
age, and impaired tissue repair. defines it as a wide range of new, returning, or have been examined in PASC versus nonPASC

Merad et al., Science 375, 1122–1127 (2022) 11 March 2022 4 of 6


C O VI D -19: 2 Y EA R S ON

convalescent individuals. Studies of individuals and shut down the production of pathogenic rapidly glean critical insights into the role of
with confirmed COVID-19 with or without PASC cytokines or reprogram pathogenic lympho- the immune system in contributing to both
found no difference in anti-S Ab levels during cytes. Evaluation of immune responses in PASC protection and pathogenesis in COVID-19. Such
the 8 months of study and no differences in the patients to vaccines is needed to reveal the insights will undoubtedly help us prepare for the
initial PCR Ct value for viral RNA (80, 81). underlying mechanism of protection. next pandemic, just as decades of previous im-
Other studies found that anti-N, -M, and -S CD8 munological research led to our current COVID-
T effector memory (TEM) cells and TEM cells Concluding remarks and future directions 19 vaccines. However, many challenges remain,
that reexpress CD45RA (TEMRA) in PASC patients The COVID-19 pandemic has wrought and our progress in ending this pandemic is
were reduced compared with those of conva- massive disruption and resulted in the loss threatened by inequitable distribution of vac-
lescent individuals, whereas anti-N T follicular of countless lives; however, there have been cines and the rise of variants that are less
helper cells (TFH) and anti-N IgG levels were silver linings. The particularly rapid develop- susceptible to vaccination and prior-infection–
elevated in blood circulation (82). These studies ment of highly efficacious vaccines is foremost mediated immunity. As infections continue to
suggest a possible persistence of viral antigens among these and has established a playbook occur, there remains a need for new therapeutics
driving immune stimulation. for the response to future pandemics. One and hence a need for a better understand-
As discussed above, autoAbs have been de- comforting prospect is the degree to which ing of the pathophysiology of COVID-19. In
tected in acute COVID-19 and evidence is em- advances in our understanding and treat- addition to treating acute infections, there is a
erging that autoAbs at the time of diagnosis were ment of COVID-19 have been aided by an dire need to better understand and develop
correlated with PASC (79). A study of 31 patients unprecedented degree of scientific coopera- treatments for individuals with PASC. An-
suffering from different PASC symptoms— tion. Free sharing of data has allowed us to other threat is the amount of misinformation
including neurological and cardiovascular and erroneous theories about the pande-
symptoms—revealed that in all 31 patients, mic, vaccines, and therapeutic efforts that
2 to 7 different GPCR-functional autoAbs have been circulating in social media,
(fAAbs) were present, acting as receptor some unfortunately introduced by scien-
agonists. Some of those GPCR-fAABs ac- tists. More than ever, interdisciplinary and
tivated their target receptors, causing a integrative approaches to scientific col-
positive chronotropic effect in neonatal Severe Asymptomatic or laboration and fighting misinformation
rat cardiomyocytes, whereas others caused COVID mild COVID are necessary to tackle these and other
a negative chronotropic effect (83). AutoAbs Autoimmunity Dysbiosis challenges that lie ahead.
to GPCR have been associated with num-
erous diseases of the cardiovascular, pul- Bacteria REFERE NCES AND N OT ES
monary, and the central nervous system 1. W. Tan et al., A Novel Coronavirus Genome Identified in
in addition to autoimmune conditions (84)

Downloaded from https://www.science.org on March 16, 2022


a Cluster of Pneumonia Cases - Wuhan, China
and may play a pathological role in me- 2019-2020. China CDC Weekly 2, 61–62 (2020).
2. W. J. Wiersinga, A. Rhodes, A. C. Cheng, S. J. Peacock,
diating PASC symptoms. H. C. Prescott, JAMA 324, 782–793 (2020).
Emergent results suggest that COVID-19 3. L. Mao et al., JAMA Neurol. 77, 683–690 (2020).
Viruuuss 4. Y. T. Chen et al., Crit. Care 24, 346 (2020).
vaccines may have an effect on PASC
5. R. Mao et al., Lancet Gastroenterol. Hepatol. 5, 667–678 (2020).
symptoms in a subset of individuals. A Hypotheses 6. B. Long, W. J. Brady, A. Koyfman, M. Gottlieb, Am. J.
prospective case control study using self- Emerg. Med. 38, 1504–1507 (2020).
reported data found that two doses of 7. K. Qi et al., Medicine 100, e25230 (2021).
8. E. A. Madden, M. S. Diamond, Curr. Opin. Virol. 52, 30–38
vaccines before SARS-CoV-2 infection (2021).
(n = 906) reduced the risk of prolonged 9. Y. Kasuga, B. Zhu, K.-J. Jang, J.-S. Yoo, Exp. Mol. Med.
symptoms after 28 days compared with 53, 723–736 (2021).
10. D. Blanco-Melo et al., Cell 181, 1036–1045.e9 (2020).
unvaccinated matched controls (n = 906)
11. I.-E. Galani et al., Nat. Immunol. 22, 32–40 (2021).
(85). Analysis of existing data on a 1296- 12. J. Hadjadj et al., Science 369, 718–724 (2020).
patient cohort with PASC emulated a 1:1 13. B. Sposito et al., Cell 184, 4953–4968.e16 (2021).
matched (vaccinated: unvaccinated) trial 14. C. Lucas et al., Nature 584, 463–469 (2020).
Viral reservoir/ Tissue damage 15. J. S. Lee et al., Sci. Immunol. 5, eabd1554 (2020).
to determine the effect of vaccination in viral remnants 16. B. Israelow et al., J. Exp. Med. 217, e20201241 (2020).
PASC patients, and found that vaccina- 17. I. Sánchez-Cerrillo et al., medRxiv
tion ~38 days postinfection significantly 2020.2005.2013.20100925 [Preprint] (2020); doi: 10.
1101/2020.05.13.20100925.
reduced the risk of prolonged symptoms 18. M. Witkowski et al., Nature 600, 295–301 (2021).
120 days postinfection (86). Even after in- 19. M. Liao et al., Nat. Med. 26, 842–844 (2020).
fection, vaccination within the first 12 weeks Post-acute sequelael of SARS 20. T. M. Delorey et al., Nature 595, 107–113 (2021).
CoV-2 infection (PASC) 21. A. J. Wilk et al., Nat. Med. 26, 1070–1076 (2020).
was associated with a reduced risk of de- 22. R. Zhou et al., Immunity 53, 864–877.e5 (2020).
Interferons
veloping Long Covid on the basis of a ret- 23. D. Mathew et al., Science 369, eabc8511 (2020).
Inflammatory cytokines
rospective analysis of the medical history 24. A. J. Wilk et al., J. Exp. Med. 218, e20210582 (2021).
Lymphocyte activation and dysregulation 25. K. A. Earle et al., Vaccine 39, 4423–4428 (2021).
of 240,648 SARS-CoV-2–infected persons
ILLUSTRATION: V. ALTOUNIAN/SCIENCE

Chronic myeloid cell activation 26. D. S. Khoury et al., Nat. Med. 27, 1205–1211 (2021).
(87). How exactly the vaccine prevents or 27. D. Corti, L. A. Purcell, G. Snell, D. Veesler, Cell 184, 3086–3108
treats PASC is currently unclear. It is pos- Fig. 2. Immunology of PASC. A fraction of COVID-19 patients with (2021).
sible that the anti-S Abs and T cells elicited either severe or mild COVID-19 develop a variety of new, recurring, or 28. T. Zohar, G. Alter, Nat. Rev. Immunol. 20, 392–394 (2020).
29. L. Piccoli et al., Cell 183, 1024–1042.e21 (2020).
by the vaccines promote clearance of resi- ongoing symptoms and clinical findings 4 or more weeks after 30. T. N. Starr et al., Cell 182, 1295–1310.e20 (2020).
dual antigens or viral particles, eliminat- infection. Analyses of immune responses in people with PASC reveal 31. M. McCallum et al., Science 373, 648–654 (2021).
ing the cause of chronic inflammation. key inflammatory cytokines and cellular activation phenotypes that are 32. D. Pinto et al., Science 373, 1109–1116 (2021).
33. Z. Wang et al., Nature 595, 426–431 (2021).
It is also possible that vaccine-induced cy- significantly elevated over nonPASC convalescent controls. Further 34. C. Lucas et al., Nature 600, 523–529 (2021).
tokines act on autoreactive lymphocytes studies are needed to identify the drivers of PASC pathophysiology. 35. S. A. Kemp et al., Nature 592, 277–282 (2021).

Merad et al., Science 375, 1122–1127 (2022) 11 March 2022 5 of 6


36. C. Rydyznski Moderbacher et al., Cell 183, 996–1012.e19 (2020). 61. N. Kaneko et al., Cell 183, 143–157.e13 (2020). 82. L. Visvabharathy et al., medRxiv 2021.2008.2008.21261763
37. K. McMahan et al., Nature 590, 630–634 (2021). 62. J. Schulte-Schrepping et al., Cell 182, 1419–1440.e23 (2020). [Preprint] (2021).
38. A. Grifoni et al., Cell 181, 1489–1501.e15 (2020). 63. A. Silvin et al., Cell 182, 1401–1418.e18 (2020). 83. G. Wallukat et al., J. Transl. Autoimmun. 4, 100100 (2021).
39. N. Le Bert et al., Nature 584, 457–462 (2020). 64. Y. Zuo et al., Sci. Transl. Med. 12, eabd3876 (2020). 84. M. A. Skiba, A. C. Kruse, Trends Pharmacol. Sci. 42, 135–150
40. J. S. Low et al., Science 372, 1336–1341 (2021). 65. C. Franke et al., Brain Behav. Immun. 93, 415–419 (2021). (2021).
41. A. Sette, S. Crotty, Nat. Rev. Immunol. 20, 457–458 (2020). 85. M. Antonelli et al., Lancet Infect. Dis. (2021).
66. S. Chakraborty et al., Nat. Immunol. 22, 67–73 (2021).
42. J. J. Guthmiller, P. C. Wilson, Science 370, 1272–1273 86. V.-T. Tran, E. Perrodeau, J. Saldanha, I. Pane, P. Ravaud, SSRN
67. E. J. Williamson et al., Nature 584, 430–436 (2020).
(2020). [Preprint] (2021); http://doi.org/10.2139/ssrn.3932953.
68. T. Takahashi et al., Nature 588, 315–320 (2020).
43. L. Swadling et al., Nature 601, 110–117 (2022). 87. M. A. Simon, R. D. Luginbuhl, R. Parker, medRxiv
69. NIH, Coronavirus Disease 2019 (COVID-19) Treatment
44. K. W. Ng et al., Science 370, 1339–1343 (2020). 2021.2011.2017.21263608 [Preprint] (2021); doi: 10.1101/2021.
Guidelines (2022) www.covid19treatmentguidelines.nih.gov).
45. H. L. Dugan et al., Immunity 54, 1290–1303.e7 (2021). 11.17.21263608.
70. D. Groff et al., JAMA Netw. Open 4, e2128568–e2128568 (2021).
46. B. S. Graham, K. Modjarrad, J. S. McLellan, Curr. Opin. 71. S. Bliddal et al., Sci. Rep. 11, 13153 (2021).
Immunol. 35, 30–38 (2015). AC KNOWLED GME NTS
72. D. Massey et al., medRxiv 2021.2007.2021.21260391 [Preprint]
47. D. M. Altmann, R. J. Boyton, Science 375, xxx–xxx (2022). (2021); doi: 10.1101/2021.07.21.21260391. We thank A. Lanzavecchia and D. Corti for helpful discussions.
48. D. A. Collier et al., Nature 593, 136–141 (2021). Due to space and reference limitations, we apologize for not being
73. C. Daniel et al., version 1, Research Square [Preprint] (2021);
49. D. Zhou et al., Cell 184, 2348–2361.e6 (2021). able to cite all relevant studies in the field. Funding: Authors
doi: 10.1101/10.21203/rs.3.rs-1139035/v1.
50. P. Wang et al., Nature 593, 130–135 (2021). acknowledge support received from P30 CA196521-05S2 (NCI
74. C. Gaebler et al., Nature 591, 639–644 (2021).
51. E. Cameroni et al., Nature (2021) COVID) (to M.M.), R01AI157488 (to A.I.), Fast Grant (George
75. C. C. L. Cheung et al., Gut 71, 226–229 (2021).
52. R. Channappanavar et al., Cell Host Microbe 19, 181–193 (2016). Mason University) (to M.M., C.B., and to A.I.), the Gates Foundation
76. M. J. Peluso et al., medRxiv 2021.2007.2009.21260287 (to M.M., A.I., and C.B.), and the Howard Hughes Medical Institute
53. L. Perico et al., Nat. Rev. Nephrol. 17, 46–64 (2021).
[Preprint] (2021); doi: 10.1101/2021.07.09.21260287. (to A.I.). Author contributions: M.M., C.B., F.S., and A.I. wrote
54. M. S. Xydakis et al., Lancet Neurol. 20, 753–761 (2021).
55. A. E. Livanos et al., Gastroenterology 160, 2435–2450.e34 77. C. Schultheiß et al., medRxiv 2021.11.16.21266391 [Preprint] the first draft, prepared figures and tables, and revised the
(2021). 2021); doi: 10.1101/2021.11.16.21266391. manuscript. Competing interests: C.B. serves as a consultant for
56. Q. Zhang et al., Science 370, eabd4570 (2020). 78. C. Phetsouphanh et al., Nat. Immunol. 23, 210–216 Catamaran Bio, DeepCell, Inc., Immunebridge, and Revelation
57. P. Bastard et al., Science 370, eabd4585 (2020). (2022). Biosciences. A.I. serves as a consultant for RIGImmune, Xanadu
58. S. E. Chang et al., Nat. Commun. 12, 5417 (2021). 79. Y. Su et al., Cell (2022). Bio, BlueWillow Biologics, and Revelar Biotherapeutics.
59. E. Y. Wang et al., Nature 595, 283–288 (2021). 80. C. Pereira et al., J. Infect. Dis. 223, 1671–1676 (2021).
60. P. Bastard et al., Sci. Immunol. 6, eabl4340 (2021). 81. H. Fang et al., Viruses 13, 916 (2021). 10.1126/science.abm8108

Downloaded from https://www.science.org on March 16, 2022

Merad et al., Science 375, 1122–1127 (2022) 11 March 2022 6 of 6


The immunology and immunopathology of COVID-19
Miriam MeradCatherine A. BlishFederica SallustoAkiko Iwasaki

Science, 375 (6585), • DOI: 10.1126/science.abm8108

View the article online


https://www.science.org/doi/10.1126/science.abm8108
Permissions
https://www.science.org/help/reprints-and-permissions

Downloaded from https://www.science.org on March 16, 2022

Use of this article is subject to the Terms of service

Science (ISSN ) is published by the American Association for the Advancement of Science. 1200 New York Avenue NW, Washington, DC
20005. The title Science is a registered trademark of AAAS.
Copyright © 2022 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim
to original U.S. Government Works

You might also like