key: cord-0955101-q1mcexa3 authors: Peng, Xiaorong; Ouyang, Jing; Isnard, Stéphane; Lin, John; Fombuena, Brandon; Zhu, Biao; Routy, Jean-Pierre title: Sharing CD4+ T Cell Loss: When COVID-19 and HIV Collide on Immune System date: 2020-12-15 journal: Front Immunol DOI: 10.3389/fimmu.2020.596631 sha: ee2b7e2df161f2d65267f51165ddcffbeb18c322 doc_id: 955101 cord_uid: q1mcexa3 COVID-19 is a distinctive infection characterized by elevated inter-human transmission and presenting from absence of symptoms to severe cytokine storm that can lead to dismal prognosis. Like for HIV, lymphopenia and drastic reduction of CD4+ T cell counts in COVID-19 patients have been linked with poor clinical outcome. As CD4+ T cells play a critical role in orchestrating responses against viral infections, important lessons can be drawn by comparing T cell response in COVID-19 and in HIV infection and by studying HIV-infected patients who became infected by SARS-CoV-2. We critically reviewed host characteristics and hyper-inflammatory response in these two viral infections to have a better insight on the large difference in clinical outcome in persons being infected by SARS-CoV-2. The better understanding of mechanism of T cell dysfunction will contribute to the development of targeted therapy against severe COVID-19 and will help to rationally design vaccine involving T cell response for the long-term control of viral infection. An outbreak of an unknown infectious pneumonia occurred in Wuhan, China, in December 2019 (1) . The pathogen of the disease was quickly identified as a novel coronavirus coined severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the disease was named coronavirus disease-19 (COVID-19) by the WHO (2) (3) (4) . The virus has since caused more than 48 million confirmed cases and over 1.2 million deaths worldwide by November, 2020 (5) . The majority of individuals with COVID-19 have mild clinical presentation with or without flu-like symptoms including dry cough, fever, a runny nose, fatigue, muscle pain and diarrhea. Some cases can evolve into acute respiratory distress syndrome, septic shock, coagulation dysfunction, and multiorgan failure (1, 6, 7) . The severity of the disease is influenced by factors such as older age, obesity and metabolic syndrome (8, 9) . Acute infection with SARS-CoV-2 is associated with lymphopenia in approximately 80% of patients (6, (10) (11) (12) (13) (14) (15) (16) (17) (18) (19) (20) (21) . Furthermore, lymphopenia with the suppression of B, helper (CD4+) and cytotoxic (CD8+) T cell function, is an indicator of a poor clinical outcome (10-15, 17-19, 21-27) . It is likely that lymphopenia delays viral clearance, favoring macrophage stimulation and the accompanying cytokine storm, leading to organ dysfunction (7, 15, 18, 19, 21, 23, 24, 26, 28, 29) . Apart from SARS-CoV-2, other viruses-including SARS coronavirus, measles virus, avian influenza virus H5N1, swine foot-and-mouth disease virus, respiratory syncytial virus and human immunodeficiency virus (HIV)-are associated with lymphopenia (30) . Among them, HIV can cause an wellknown lymphopenia-associated disease acquired immune deficiency syndrome (AIDS) (31) . The acute phase of HIV infection is characterized by a substantial drop in peripheral CD4+ T cell counts, while during the chronic phase, a slower and persistent decline of these CD4+ T cells is associated with the development of AIDS. Antiretroviral therapy (ART) rapidly suppresses HIV replication, and the number of CD4+ T cell counts recovers, preventing AIDS. However, systemic immune activation persists in those people even after years of ART (32) , and is characterized by increased proinflammatory mediators and low CD4/CD8 ratio (33) , combined with exhausted and senescent T cells. Systemic immune activation is also associated with non-infectious comorbidities, such as cardiovascular diseases, neurocognitive disorders and cancers. CD4+ T cells orchestrate the response to acute and chronic viral infections by coordinating the immune system. These cells activate multiple cells of the innate immune system, as well as B cells, cytotoxic CD8+ T cells, and non-immune cells. CD4+ T cells also play a key role for the establishment of long-term cellular and humoral antigen specific immunity, which is the basis of life-long protection for many viral infections and vaccines (34, 35) . Both HIV-1 and SARS-CoV-2 have distinct virological characteristics while sharing CD4+ T cell lymphopenia. In this review, we critically assessed the possible mechanisms and the potential influence of CD4+ T cell lymphopenia in acute and chronic viral infections. We also discuss host characteristics and hyper-inflammatory response in these two dramatic viral infections and the impact of COVID-19 infection in people living with HIV (PLWH). The acute phase of HIV infection is characterized by a substantial drop in peripheral CD4+ T cell counts while in the chronic phase, a continued decline of CD4+ T cells is associated with the development of AIDS ( Figure 1 ). In contrast, expansion of CD8+ T cells is observed which is driven mainly by an exhausted cytotoxic response toward HIV, leading to an inversed CD4/CD8 ratio. Despite ART, PLWH still present persistent immune activation and inflammation. The expressions of CD38 and HLA-DR as well as programmed death -1 (PD-1) are biomarkers of activated T cells, contributing to T cell exhaustion (36) . Exhausted virusspecific CD4+ T cells express PD-1 at elevated levels correlating with disease progression, viral loads and reduced CD4+ T cell count (37) . Compared to healthy controls, in both acutely and chronically PLWH, the absolute number of regulatory T cells (Tregs) in the circulating blood is decreased, however the percentage of Tregs in chronic infection is increased (38, 39) , further contributing to T cell dysfunction. Gut CD4+ T cells with a mucosa protective Th17 function are rapidly depleted (40) contributing to mucosal barrier dysfunction, leading to increase microbial translocation and systemic immune activation (41) . Despite the decreased CD4+ T cell subgroup, both cell number and relative percentage of circulating T follicular helper (Tfh) cells increased in the blood during the chronic phase of HIV infection (42, 43) . Tfh cells provide help to B cells in germinal center of secondary lymphoid organs and are central to the generation of efficient neutralizing and non-neutralizing antibody responses in HIV infection and will be essential in generating an effective vaccine (44) . Expansion and altered features of HIV-specific and non-HIV specific circulating Tfh cells do not improve during ART and may be driven by persistent HIV antigen expression (45) . Viral suppression by ART resulted with a reduction in the expression of genes associated with Tfh cells compared to viremic phase, which is accompanied by persistently low expression of genes associated with Th17 cells compared to persons who spontaneously control viremia (46) . Lymphocytopenia is a hallmark of patients with severe COVID-19 (6, 10-15, 17-19, 21) and is associated with poor clinical outcomes. The CD4+ lymphocyte count dynamic during mild and severe COVID-19 is shown in Figure 1 . Helper CD4+ T cells are important in mediating protective humoral immunity by stimulating B cells to produce virus-specific antibodies. On the other hand, CD8+ T cells are responsible for the elimination of infected cells, mainly through the production of perforin and granzyme, and are key players in controlling different types of viruses through the secretion of cytokines. Both CD4+ and CD8+ T cell counts are reduced in severe COVID-19 (10-15, 17-19, 21-27) . Similarly, reduced B cell counts are also observed in severe COVID-19 (14, 23) . Moreover, within the CD4+ T cell subset, decreased numbers of effector memory T cells (CD45RO+) and Tregs (CD25+CD127low) were noted, while the proportion of naive T cells (CD45RA+) increased (16) . The frequency of Tregs, which are responsible for the maintenance of immune homeostasis by suppressing activation and proinflammatory functions, was very low in severe cases. In addition, relative increased recirculation of activated CXCR5 +PD-1 high CD4+ Tfh cells is observed in severe COVID-19. CD4+ T cells in COVID-19 are activated as characterized by expression of cellular markers like HLA-DR, CD25, CD38 and Ki-67 (47) . T cell exhaustion based on increased inhibitory markers such as PD-1 and TIM-3 receptor on peripheral T cells has also been reported (47) (48) (49) . Studies have demonstrated that decreases in polyfunctionality (multiple cytokine secretion) and cytotoxicity of T cells were correlated with disease progression (21, 49) . Conversely to HIV, a study demonstrated an increase in the number of Th17 cells in the peripheral blood in COVID-19 patients (50) . In hospitalized patients compared to non-hospitalized patients, Mathew et al. found increased proportion of cytotoxic follicular helper cells and cytotoxic T helper cells responding to SARS-CoV-2 and reduced proportion of SARS-CoV-2-reactive Treg cells (47) . Elevated SARS-CoV-2specific CD4+ and CD8+ T cells were each associated with milder disease, fostering important roles for both CD4+ and CD8+ T cells in protective immunity in COVID-19 (51) . Furthermore, absence of these virus-specific cells leads to uncoordinated antigen-specific immune responses and failure to control COVID-19, predominantly in older individuals with low naïve CD4+ T cells. Similarly, PLWH are not able to mount an effective HIV-specific CD4+ and CD8+ T cell responses with the exception of HIV controllers who can maintain undetectable or low levels of viremia despite not being on ART (46, 52, 53) . Features of different peripheral blood cell types in PLWH and severe COVID-19 are shown in Figure 2 . Depletion of gut CD4+ T cells will be followed by disruption of the tight junctions, and cell death of intestinal epithelium. Epithelial gut damage leads to both an imbalance of the intestinal microbiota composition (dysbiosis) and the release of bacterial products in the circulation (microbial translocation), participating in chronic immune activation and inflammation (54, 55) . Apart from relevant metabolic functions for the host homeostasis, the gut microbiota exerts protective actions against pathogenic colonization of bacteria and viruses, which could be at least partially attributed to their role in educating and strengthening the immune system (56) . The triad gut microbiota dysbiosis-immune hyper-response-inflammation is involved in both HIV and COVID-19 pathogenesis (57) . Within a few weeks of HIV infection, the virus begins a massive assault on the gut, which undergoes a significant depletion of CD4+ T cells with Th17 function (58) . PLWH have an altered microbiota composition with an increase of proinflammatory and potentially pathogenic bacteria as well as a decrease of beneficial bacteria (59, 60) . Gut damage allows microbial translocation, a cause of systemic immune activation in chronic HIV which is usually determined by measuring plasma levels of markers of microbial translocation such as lipopolysaccharide (LPS) and (1!3)-b-D-Glucan (BDG), all of which are elevated in PLWH, even those on ART (61, 62) . Previous studies have shown that LPS and BDG were associated with disease progression, lower CD4+ T cell count, and induce immune activation (61) (62) (63) . Over 60% of patients with COVID-19 report evidence of gastrointestinal symptoms, such as diarrhea, nausea and vomiting (64) . There is direct evidence that SARS-CoV-2 can replicate in intestinal cells (65) . Moreover, many viral infections, including influenza, drive changes in the gut and lung microbiota with viral-mediated changes in the gut including dysbiosis and increased permeability (66) . Indeed, recent studies found some differences in gut microbial features and related metabolites in SARS-CoV-2 infection (67) . More attention should be directed to gut dysbiosis and microbial translocation in the contribution to severe COVID-19. Examination of plasma cytokines of acute HIV infection revealed that interferon (IFN)-a was the first cytokine to be increased within a few days after detection of viremia, followed by tumor necrosis factor a (TNF-a), IFN-g, and interleukin (IL)-12 (68) . Initiation of ART during Fiebig stages I-II can abrogate the HIVinduced cytokine storm (69) . Elevation of IFN-a, IFN-g, monocyte chemoattractant protein (MCP)-1, soluble IL-2 receptor (sCD25), IL-6 and IL-8 was seen in chronicallyinfected untreated individuals (63, 70, 71) . Initiation of ART significantly reduces plasma levels of inflammatory cytokines, markers of inflammation and monocyte activation, without normalization compared to HIV-uninfected individuals (72) . Similarly, in COVID-19 patients, elevation of inflammatory cytokines was also observed. In severe cases, elevations of TNF-a, IFN-g, IL-2R, IL-6, IL-8, and IL-10 were detected (7, 15, 18, 19, 21, 23, 24, 26, 28, 29) . However, a highly impaired interferon (IFN) type I response was observed, characterized by no IFN-b and low IFN-a production and activity (73) . Furthermore, studies have found increased production of proinflammatory cytokines and chemokines, IL-2, IL-7, IL-10, granulocyte colony-stimulating factor (G-CSF), CXCL-10/IP-10, TNF-a and macrophage inflammatory protein (MIP)-1a in intensive care unit (ICU) patients compared with non-ICU patients (7) . In addition, IL-6 levels were considered as a biomarker of disease severity and mortality (28, 29) and ongoing clinical trials are assessing IL-6 blockade to improve outcome in COVID-19 patients (74) . A pre-existed low-level inflammation and leaky gut in type 2 diabetes mellitus (T2DM) may be associated with higher COVID-19 mortality (75, 76) . Retrospective studies have shown a reduction in mortality in metformin users compared with non-users among patients with T2DM hospitalized for COVID-19 (77) . The potential effects of metformin in COVID-19 could be through inhibition of the mTOR pathway and prevention of immune hyperactivation (78) . Reduced production of cytokines such as TNF-a and IL-6 was seen in metformin-treated patients (79) . Furthermore, metformin may also reduce inflammation by altering the composition of gut microbiota (80, 81) . A retrospective cohort study on PLWH with diabetes mellitus showed that Metformin use was associated with improved CD4 recovery (82) . Whether metformin could be a potential treatment strategy for CD4+ T cells lymphopenia in COVID-19 need further investigation. Although ART reduced the risk of developing AIDS (83), it does not normalize inflammation that is associated with risk of non-AIDS comorbidities, including cardiovascular and metabolic diseases and neurocognitive dysfunctions (84) (85) (86) . In COVID-19, direct viral attack and systemic hyperinflammation can cause dysfunction of several organs. Postmortem analyses showed that the main damage occurred in the lungs, to the alveolar epithelial cells, hyaline membrane formation, and hyperplasia of type II pneumocytes, all components of diffuse alveolar damage (87, 88) . Nearly 20% of patients hospitalized for COVID-19 in Wuhan, China showed evidence of cardiac injury (89, 90) . More than half of COVID-19 patients hospitalized had elevated levels of enzymes indicating injury to the liver (91). In a case series of 214 patients with COVID-19, neurologic symptoms were seen in 36.4% of patients which included acute cerebrovascular events, impaired consciousness, and muscle injury (92) . The thymus supports T cell differentiation from T progenitor cells, which differentiate from hematopoietic stem cells in bone marrow, and selects mature CD3+ CD4+ and CD3+ CD8+ thymocytes (41) . Quantitative estimates indicate that healthy young (<30 year old) adults harbor about 2.2×10 11 mature CD4+ T cells (93) . Most CD3+CD4+ and CD3+CD8+ T cells reside in peripheral lymphoid organs where T and B cell responses are coordinated by antigen-presenting cells (APC). CD4+ T cell numbers are kept constant in the human body by homeostatic mechanisms including IL-7 (41) . Total CD4+ T cells may be depleted due to cell death, shortened half-life or impaired production. In addition, the proportion of circulating CD4+ cells may decrease through lymphoid tissue redistribution at sites of inflammation. A number of dynamic models have been put forth explaining HIV-mediated depletion of CD4+ T cells (94, 95) . However, CD4+ lymphopenia is poorly understood in COVID-19. Potential mechanisms and consequences of CD4+ lymphopenia in PLWH and COVID-19 are shown in Figure 3 . Early experiments done with laboratory-adapted HIV isolates in tissue culture revealed a cytopathic virus with high tropism for CD4+ T cells (96) . There is a homeostatic response by which the loss of CD4+ T cells due to HIV infection is counteracted by production of T cells; however, this balance is ultimately disrupted once the production of T cells in response to homeostasis is exhausted. This has been substantiated by quantitative image analysis of decreased numbers of CD4+ T cells and increased levels of cellular proliferation and apoptosis in PLWH (97, 98) . However, evidence showed that HIV pathogenesis cannot be solely explained by the direct viral killing hypothesis as uninfected CD4+ T cells have a shortened half-life by cellular viral contact affecting IL-7 signalization (99). Another explanation is phospholipase A2 group IB (PLA2G1B) which synergizes with the HIV gp41 envelope protein and targets the CD4+ T cell surface, leading to CD4+ T cell unresponsiveness (anergy) (100) . The question arises whether SARS-CoV-2, like HIV, can directly decrease CD4+ T cell count. ACE2 (angiotensin-converting enzyme 2) is the SARS-CoV-2 internalization receptor (101) , in concert with the host's TMPRSS2 (transmembrane protease serine 2) membrane protease that primes the coronavirus spike S protein to facilitate its cell entry (102) . ACE2 and TMPRSS2 are coexpressed in lung, heart, liver, kidney, neurons and immune cells (103) . Immune cells could potentially be infected by SARS-CoV-2, as in the case of SARS-CoV (104), with both viruses sharing the same receptor ACE2 (102) . Studies showed that SARS-CoV can infect 50% of lymphocytes in the circulation (105) , resulting in cell death by apoptosis, necrosis, or pyroptosis (106, 107) . Furthermore, under the influence of SARS-CoV, the germinal center regressed, and both T and B lymphocytes are depleted (108) . Extensive cell death of lymphocytes was observed in an autopsy study of spleens and hilar lymph nodes of six patients with COVID-19. However, the direct evidence of whether SARS-CoV-2 infects T cells is still lacking. Previous studies proposed that activated CD4+ T cells have a very short life span due to activation-induced cell death or apoptosis (109) . In HIV infection, the activation of CD4+ T cells is driven by the antigenic stimulus by HIV proteins (110) and in part by antigen-independent mechanisms through the production of inflammatory cytokines. Continuous hyperactivation of T cells may lead to accelerated consumption of naïve T cells through apoptosis or differentiation toward a memory phenotype. Elevation of inflammatory cytokines and cytokine storm was observed in COVID-19 patients. Previous studies showed that a number of inhibitory cytokines are released by infected lung macrophages or epithelial cells. These cytokines include TNF-a which causes T cell apoptosis (111) , IL-10 which is known to prevent T cell proliferation (112) , and type-I IFN which regulates lymphocyte recirculation (113) . Whether these inflammatory cytokines contribute to the loss of CD4+ T cell needs further investigation. Circulating CD4+ T cell counts are most studied due to their ease of access. However, CD4+ T cell in the blood compartment does not always reflect the composition of lymphoid organs or infected sites where CD4+ T cells are recruited. Hence, CD4+ T cell lymphopenia could be a reflection of CD4+ T cell redistribution throughout the body. Some evidence from simian immunodeficiency virus (SIV) macaque models indicates CD4+ T cell redistribution from the peripheral blood to lymph nodes and the gut (114) . When blood levels of CD4+ lymphocytes begin to drop significantly, these cells often increase in number in the lymph nodes (115) . This suggests that the loss of CD4+ T cells in the blood can in part be explained by an enhanced homing of CD4+ lymphocytes into the lymph nodes. Furthermore, CD62L, the receptor for lymph node homing, could be unregulated after infection with HIV (116) . After the initiation of effective antiretroviral therapy, decreased levels of adhesion molecules like VCAM-1 and ICAM-1, which mediate lymphocyte sequestration into lymphoid tissue, were associated a rapid increased of CD4 T cells and decreased LN size (117) . SARS-CoV-2 prefentially infects and destroys alveolar epithelial cells that may in turn trigger the production or the overproduction by macrophages of pro-inflammatory cytokines and chemokines (including interleukin-6 (IL-6), IL-8, CXCL10/ IP-10, CCL3/MIP1a, CCL4/MIP1b) (118) . Secretion of such cytokines and chemokines attracts immune cells, notably monocytes and T lymphocytes, from the blood into the infected site, which may explain the circulating lymphopenia. Additionally, the first autopsy of a patient with COVID-19 revealed an accumulation of mononuclear cells (monocytes and T cells) in the lungs, coupled with low levels of hyperactive T cells in the peripheral blood (88) . Furthermore, anti-IL-6 immediately reversed lymphopenia favoring tissue redistribution in patients having multicentric Castleman disease, a condition characterized by an enhanced level of IL-6 (119). Animal models and future clinical trials will help decipher the mechanism responsible for SARS-CoV-2 associated lymphopenia. Several case reports assessed the influence of COVID-19 in PLWH (120) (121) (122) (123) . In a case series of 33 PLWH patients with COVID-19, three out of 32 patients with documented outcome died (9%). However, 91% of the patients recovered and 76% have been classified as mild cases, indicating that there is no excess morbidity and mortality among PLWH with symptomatic COVID-19 compared to COVID-19 HIV-negative patients (123) . In a study in Wuhan, there were 8 COVID-19 out of 1174 investigated HIV/AIDS patients. The authors reported absence of influence of sex, CD4+ T cells counts, HIV viral load, or ART regimen associated with the occurrence of COVID-19, only older age was associated with COVID-19 infection (124) . A compromised immune system with a lower CD4+ T cells counts and elevated interferon levels in HIV infection might reduce clinical symptoms of COVID-19. There is a hypothesis that a lower active immune status might protect the human body from a virus-induced cytokine storm, such as SARS and MERS (125) . Some ART medications (lopinavir/ritonavir, ritonavir, darunavir, and dolutegravir), were screened for anti-SARS-CoV-2 replication activity and were initially used to treat COVID-19 (126) . However, clinical trials using lopinavir/ ritonavir, a protease inhibitor that could suppress SARS-CoV-2 replication in vitro, had no impact on COVID-19 outcome (127) . Another drug is tenofovir (TDF), a nucleoside analog of remdesivir, which can inhibit SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) activity in vitro and shorten the time to recovery in adults who were hospitalized with COVID-19 and had evidence of lower respiratory tract infection (128) . In a cohort study with 77 590 HIV-positive persons receiving ART, the result showed that HIV-positive patients receiving TDF/ Emtricitabine (FTC) had a lower risk for COVID-19 and related hospitalization than those receiving other therapies (129) . These findings warrant further investigation in healthy individuals taking these two drugs for HIV preexposure prophylaxis studies and randomized trials in persons with and without HIV. T CELLS LYMPHOPENIA IL-7 IL-7 levels are known to be inversely correlated with CD4+ T cell counts in patients with HIV/AIDS, and is likely associated with a homeostatic response (130) . IL-7 is essential to B and T cell lymphopoiesis in the bone marrow. Clinical studies showed that recombinant IL-7 treatment increased the number of naive and memory CD4+ and CD8+ T cells while conserving T cell functions (131, 132) . Several clinical trials are currently under way to evaluate the efficacy of IL-7 to improve clinical outcomes in lymphopenic patients with COVID-19 (NCT04407689, NCT04379076, NCT04442178 and NCT04442178). IL-2 is a potent mitogen and growth factor in antigen-stimulated CD4+ T cells (133) . IL-2 has been studied in HIV and has been shown to increase CD4+ T cells counts (134, 135) . IL-2 levels in the peripheral blood were increased in severe COVID-19 cases compared to mild cases (7, 15, 18, 19, 21, 23, 24, 26, 28, 29) . Whether IL-2 can be used to improve CD4+ T cell lymphopenia in COVID-19 patients should be carefully considered. The efficacy of low-dose IL-2 administration is under evaluation in patients with SARS-CoV2-related acute respiratory distress syndrome in a randomized controlled trial (NCT04357444). There are still some knowledge gaps about CD4+ T cell loss in PLWH and COVID-19. Firstly, the dynamic of change in CD4+ T cell is difficult to be compared, especially as HIV induces both an acute and chronic disease state. Secondly, the data in COVID-19 are limited. New studies need to be conducted to learn more about this new disease, and lessons from studies on HIV infection and care of PLWH could definitely help designing new therapeutic tools. Both HIV-1 and SARS-CoV-2 infection share CD4+ T cell loss in association with disease outcome and immunodeficiency. Direct attacks on CD4+ T cells, immune activation and redistribution of CD4+ T cell are contributing mechanisms in very different proportion for CD4+ T cell lymphopenia in both diseases. During the period of immunodeficiency, systemic inflammation could be fueled by leaky gut and lead to severe complications. However, when HIV meets COVID-19, no increase in the occurrence of COVID-19 and no excess morbidity and mortality among PLWH with symptomatic COVID-19 has been reported. IL-7 and IL-2 were previously used to increase CD4+ T cell counts in HIV-1 infection, however, no improvement in their function were reported. Despite this, the short-term effect for COVID-19 is under investigation. As CD4+ T cells orchestrate immune responses, proper CD4+ T cell function is required for effective vaccine responses. Hence, anti-SARS-CoV-2 antibodies and CD4 responses should be studied in order to develop long-term efficiency vaccine formulation. Overall, experience in HIV clinical management and past clinical trials represent a special use case for innovative studies aiming at increasing CD4+ T cell function and reducing COVID-19 morbidity. XP wrote the first draft of the manuscript. JO, SI, JL, BF, and BZ provided critical revision of the manuscript. J-PR conceived and designed the manuscript. All authors contributed to the article and approved the submitted version. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study Coronaviridae Study Group of the International Committee on Taxonomy of V. The species Severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2 A pneumonia outbreak associated with a new coronavirus of probable bat origin A Novel Coronavirus from Patients with Pneumonia in China An interactive web-based dashboard to track COVID-19 in real time Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China Clinical features of patients infected with 2019 novel coronavirus in Wuhan Risk Factors Associated With Acute Respiratory Distress Syndrome and Death in Patients With Coronavirus Disease COVID-19 and smoking: A systematic review of the evidence Risk factors associated with disease progression in a cohort of patients infected with the 2019 novel coronavirus Epidemiological characteristics and clinical features of 32 critical and 67 noncritical cases of COVID-19 in Chengdu Suppressed T cellmediated immunity in patients with COVID-19: A clinical retrospective study in Wuhan Epidemiological and initial clinical characteristics of patients with family aggregation of COVID-19 Characteristics of peripheral lymphocyte subset alteration in COVID-19 pneumonia CD4 Loss in COVID-19 and HIV Frontiers in Immunology | www Relationships among lymphocyte subsets, cytokines, and the pulmonary inflammation index in coronavirus (COVID-19) infected patients Dysregulation of immune response in patients with COVID-19 in Wuhan, China Lymphocyte subset (CD4+, CD8+) counts reflect the severity of infection and predict the clinical outcomes in patients with COVID-19 Correlation Between Relative Nasopharyngeal Virus RNA Load and Lymphocyte Count Disease Severity in Patients with COVID-19 Complex Immune Dysregulation in COVID-19 Patients with Severe Respiratory Failure Increased expression of CD8 marker on T-cells in COVID-19 patients Clinical and immunological features of severe and moderate coronavirus disease 2019 One Case of Coronavirus Desease 2019(COVID-19) in Patient Co-nfected by HIV With a Low CD4+ The laboratory tests and host immunity of COVID-19 patients with different severity of illness Clinical features of severe pediatric patients with coronavirus disease 2019 in Wuhan: a single center's observational study Down-regulated gene expression spectrum and immune responses changed during the disease progression in COVID-19 patients Coronavirus COV-19/SARS-CoV-2 affects women less than men: clinical response to viral infection Clinical progression of patients with COVID-19 in Shanghai Diagnostic utility of clinical laboratory data determinations for patients with the severe COVID-19 Hematologic, biochemical and immune biomarker abnormalities associated with severe illness and mortality in coronavirus disease 2019 (COVID-19): a metaanalysis Significant changes of peripheral T lymphocyte subsets in patients with severe acute respiratory syndrome Albeit Reduced, Chronic Inflammation in Persons Starting Antiretroviral Therapy in Acute HIV Infection Determinants of a Low CD4/CD8 Ratio in HIV-1-Infected Individuals Despite Long-term Viral Suppression Influenza vaccine-induced human bone marrow plasma cells decline within a year after vaccination Effector and memory T-cell differentiation: implications for vaccine development Immune activation and CD8+ T-cell differentiation towards senescence in HIV-1 infection Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction Homeostasis and function of regulatory T cells in HIV/SIV infection Regulatory T cells in HIV infection: can immunotherapy regulate the regulator? Loss and dysregulation of Th17 cells during HIV infection The dynamics of CD4+ T-cell depletion in HIV disease Expansion of HIV-specific T follicular helper cells in chronic HIV infection CD4 T Cells Mediate Both Positive and Negative Regulation of the Immune Response to HIV Infection: Complex Role of T Follicular Helper Cells and Regulatory T Cells in Pathogenesis Emerging concepts on T follicular helper cell dynamics in HIV infection Persistent expansion and Th1-like skewing of HIV-specific circulating T follicular helper cells during antiretroviral therapy Altered differentiation is central to HIV-specific CD4(+) T cell dysfunction in progressive disease Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications Elevated exhaustion levels and reduced functional diversity of T cells in peripheral blood may predict severe progression in COVID-19 patients Functional exhaustion of antiviral lymphocytes in COVID-19 patients The clinical course and its correlated immune status in COVID-19 pneumonia Antigen-Specific Adaptive Immunity to SARS-CoV-2 in Acute COVID-19 and Associations with Age and Disease Severity HIV controllers maintain a population of highly efficient Th1 effector cells in contrast to patients treated in the long term Preserving HIV-specific T cell responses: does timing of antiretroviral therapy help? Circulating LPS and (1->3)-beta-D-Glucan: A Folie a Deux Contributing to HIV-Associated Immune Activation Association Between Gut Microbiota and CD4 Recovery in HIV-1 Infected Patients The interplay between the gut microbiota and the immune system The Role of Angiotensin Converting Enzyme 2 (ACE2) in Modulating Gut Microbiota, Intestinal Inflammation, and Coronavirus Infection HIV-1 selectively targets gut-homing CCR6+CD4+ T cells via mTORdependent mechanisms Dysbiosis of the gut microbiota is associated with HIV disease progression and tryptophan catabolism A compositional look at the human gastrointestinal microbiome and immune activation parameters in HIV infected subjects Circulating (1->3)-beta-D-glucan Is Associated With Immune Activation During Human Immunodeficiency Virus Infection Microbial translocation is a cause of systemic immune activation in chronic HIV infection CXCL13 as a Biomarker of Immune Activation During Early and Chronic HIV Infection SARS-CoV-2 induced diarrhoea as onset symptom in patient with COVID-19 SARS-CoV-2 productively infects human gut enterocytes Respiratory Viral Infection-Induced Microbiome Alterations and Secondary Bacterial Pneumonia Gut Microbiota Dysbiosis-Immune Hyperresponse-Inflammation Triad in Coronavirus Disease Induction of a striking systemic cytokine cascade prior to peak viremia in acute human immunodeficiency virus type 1 infection, in contrast to more modest and delayed responses in acute hepatitis B and C virus infections Association between the cytokine storm, immune cell dynamics, and viral replicative capacity in hyperacute HIV infection Changes in Inflammation and Immune Activation With Atazanavir-, Raltegravir-, Darunavir-Based Initial Antiviral Therapy: ACTG 5260s Dynamics of immune reconstitution and activation markers in HIV+ treatment-naive patients treated with raltegravir, tenofovir disoproxil fumarate and emtricitabine Inflammation, immune activation, and cardiovascular disease in HIV Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients Tocilizumab in patients with severe COVID-19: a retrospective cohort study Hypothesized mechanisms explaining poor prognosis in type 2 diabetes patients with COVID-19: a review COVID-19 and diabetes mellitus: how one pandemic worsens the other Metformin and COVID-19: From cellular mechanisms to reduced mortality Is metformin ahead in the race as a repurposed hostdirected therapy for patients with diabetes and COVID-19? Clinical Characteristics and Outcomes of Patients With Diabetes and COVID-19 in Association With Glucose-Lowering Medication Metformin effect on gut microbiota: insights for HIV-related inflammation Repurposing metformin in non-diabetic people living with HIV: Influence on weight and gut microbiota Cohort study of diabetes in HIV-infected adult patients: evaluating the effect of diabetes mellitus on immune reconstitution Immune activation and inflammation in HIV-1 infection: causes and consequences T cell activation and senescence predict subclinical carotid artery disease in HIV-infected women Fracture prevalence among human immunodeficiency virus (HIV)-infected versus non-HIV-infected patients in a large U.S. healthcare system The rising challenge of non-AIDS-defining cancers in HIV-infected patients Pathological study of the 2019 novel coronavirus disease (COVID-19) through postmortem core biopsies. Modern Pathol an Off J U States Can Pathological findings of COVID-19 associated with acute respiratory distress syndrome Association of Cardiac Injury With Mortality in Hospitalized Patients With COVID-19 in Wuhan, China Characteristics of Ocular Findings of Patients With Coronavirus Disease Liver injury in COVID-19: management and challenges Neurologic Manifestations of Hospitalized Patients With Coronavirus Disease Population biology of HIV-1 infection: viral and CD4+ T cell demographics and dynamics in lymphatic tissues Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection Immune activation set point during early HIV infection predicts subsequent CD4+ Tcell changes independent of viral load Viral dynamics in human immunodeficiency virus type 1 infection T cell depletion in HIV-1 infection: how CD4+ T cells go out of stock HIV-1 causes CD4 cell death through DNA-dependent protein kinase during viral integration Long-term kinetics of T cell production in HIV-infected subjects treated with highly active antiretroviral therapy PLA2G1B is involved in CD4 anergy and CD4 lymphopenia in HIV-infected patients Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor Influenza and SARS-coronavirus activating proteases TMPRSS2 and HAT are expressed at multiple sites in human respiratory and gastrointestinal tracts Multiple organ infection and the pathogenesis of SARS Immunological responses against SARS-coronavirus infection in humans SARS-Coronavirus Open Reading Frame-3a drives multimodal necrotic cell death Induction of apoptosis by the severe acute respiratory syndrome coronavirus 7a protein is dependent on its interaction with the Bcl-XL protein Severe acute respiratory syndrome associated coronavirus is detected in intestinal tissues of fatal cases Understanding the slow depletion of memory CD4+ T cells in HIV infection HIV-1 Nef mediates lymphocyte chemotaxis and activation by infected macrophages Interleukin-10 determines viral clearance or persistence in vivo Type I interferons directly regulate lymphocyte recirculation and cause transient blood lymphopenia Acute Simian Immunodeficiency Virus Infection Triggers Early and Transient Interleukin-7 Production in the Gut, Leading to Enhanced Local Chemokine Expression and Intestinal Immune Cell Homing An immunohistological approach to persistent lymphadenopathy and its relevance to AIDS HIV induces homing of resting T lymphocytes to lymph nodes Initial increase in blood CD4(+) lymphocytes after HIV antiretroviral therapy reflects redistribution from lymphoid tissues The trinity of COVID-19: immunity, inflammation and intervention Improvement in Castleman's disease by humanized anti-interleukin-6 receptor antibody therapy COVID-19 in patients with HIV: clinical case series Early virus clearance and delayed antibody response in a case of COVID-19 with a history of coinfection with HIV-1 and HCV Co-infection of SARS-CoV-2 and HIV in a patient in Wuhan city COVID-19 in people living with human immunodeficiency virus: a case series of 33 patients A Survey for COVID-19 Among HIV/AIDS Patients in Two Districts of Wuhan Why Aren't People Living with HIV at Higher Risk for Developing Severe Coronavirus Disease Predicting commercially available antiviral drugs that may act on the novel coronavirus (SARS-CoV-2) through a drug-target interaction deep learning model A Trial of Lopinavir-Ritonavir in Adults Hospitalized with Severe Covid-19 Remdesivir for the Treatment of Covid-19 -Final Report Incidence and Severity of COVID-19 in HIV-Positive Persons Receiving Antiretroviral Therapy : A Cohort Study Decreased interleukin 7 responsiveness of T lymphocytes in patients with idiopathic CD4 lymphopenia Cycles of Recombinant Human Interleukin 7 in HIV-Infected Patients With Low CD4 T-Cell Reconstitution on Antiretroviral Therapy: Results of 2 Phase II Multicenter Studies Effects of recombinant human interleukin 7 on T-cell recovery and thymic output in HIV-infected patients receiving antiretroviral therapy: results of a phase I/IIa randomized, placebo-controlled, multicenter study Signaling and Function of Interleukin-2 in T Lymphocytes Interleukin-2 therapy in patients with HIV infection Immunological effects of interleukin-2 therapy in human immunodeficiency virus-positive subjects The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.Copyright © 2020 Peng, Ouyang, Isnard, Lin, Fombuena, Zhu and Routy. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.