Abstract
After healing from COVID-19, patients often experience a slew of symptoms known as post COVID-19 sequelae. Despite the fact that the SARS-CoV-2 pandemic is still ongoing, post-Covid-19 syndrome is already a difficult problem to address: long-term multiorgan sequelae, while frequently described, have yet to be systematized. As a result, post-Covid-19 syndrome can have a major influence on surviving patients’ working capacity as well as their personal lives. The clinical spectrum and long-term course of this clinical entity must be better understood. Post-Covid syndrome affects a wide spectrum of individuals (16–87%), with pneumological and cognitive symptoms being the most common. Pulmonary fibrosis was the most common organic consequence seen in post-Covid patients. In conclusion, post-Covid-19 syndrome can have a major impact on the health of survivors. Working-age patients should seek rehabilitation and follow-up in interdisciplinary rehabilitation programmes. Given the pandemic’s global extent, it’s obvious that COVID-19-related healthcare demands will continue to climb for the foreseeable future. For COVID-19 survivors’ long-term mental and physical health, present outpatient infrastructure will be utilised, scalable healthcare models will be built, and cross-disciplinary collaboration will be required.
Introduction
Coronavirus disease 2019 (COVID-19), caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is one of the most devastating infectious disease in recent times. SARS-CoV-2 was declared a global pandemic by the World Health Organization (WHO) [1], that initially started from Wuhan, China, and spread extremely rapidly, making its way to over 180 countries around the world. SARS-CoV-2 is an enveloped beta-coronavirus. There are seven species of beta-coronaviruses can cause human infections. Three out of these seven human infections, namely severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and COVID-19 are potentially fatal [2]. The angiotensin-converting enzyme 2 (ACE2) receptor in human cells acts as the functional receptor for SARS-CoV-2, facilitating its entry into the cell and plays a critical role in COVID-19 pathogenesis [3]. ACE2 receptor is abundantly expressed on smooth muscle cells and endothelia in virtually all organs [4] (Table 1).
Summary of post-acute COVID-19 by organ system.
Sr no. | System | Manifestations |
---|---|---|
1 | Respiratory | Shortness of breath, dry cough, haemoptysis, dyspnoea, decreased exercise capacity, hypoxia, reduced diffusion capacity and restrictive pulmonary physiology |
2 | Hematologic | Thromboembolic events |
3 | Cardiovascular | Palpitations, dyspnoea, chest pain, long-term sequelae includes increased cardiometabolic demand, myocardial fibrosis or scarring (detectable via cardiac magnetic resonance imaging (MRI)), arrhythmias, tachycardia and autonomic dysfunction |
4 | Neuropsychiatric | Fatigue (most common sequelae), myalgia, headache, dysautonomia, cognitive impairment (brain fog), anxiety, depression, sleep disturbances, post-traumatic stress disorder (PTSD), changes in concentration, impaired memory, persistent muscle weakness and myalgias, headaches, sleep disorders, dizziness, impairment in smell (anosmia) and taste (ageusia), new onset status epilepticus, stroke, acute inflammatory demyelinating polyneuritis (AIDP), autonomic dysfunction such as orthostatic symptoms and non-specific sensory complaints such as paraesthesia, numbness, tingling in limbs |
5 | Renal | Nocturia (frequent urination during night which disturbs sleep), increased froth in urine, anorexia (loss of appetite), headache, swelling on leg, orthopnoea (breathing difficulty during lying down), new onset acute kidney injury (AKI) rapid progression of pre-existing chronic kidney disease (CKD), new onset glomerular disease – proteinuria, haematuria and renal dysfunction |
6 | Endocrine | New or worsening control of existing diabetes mellitus, subacute thyroiditis, bone demineralization, suspected hypothalamic–pituitary–adrenal axis suppression or hyperthyroidism |
7 | Gastrointestinal and hepatobiliary | Diarrheal, dyspepsia, nausea/vomiting, abdominal pain, dysgeusia, loss of appetite, hematemesis/melena, dysphagia, G.E.R.D, alteration of the gut microbiome, including enrichment of opportunistic organisms and depletion of beneficial commensals, prolonged viral faecal shedding even after negative nasopharyngeal swab testing |
8 | Dermatologic | Hair loss is the most predominant symptom and skin rash |
9 | MIS-C | Children >7 years and <21 years old with fever, elevated inflammatory markers, multiple organ dysfunction, current or recent SARS-CoV-2 infection. Cardiovascular (coronary artery aneurysm) and neurologic (headache, encephalopathy, stroke and seizure) complications can occur |
10 | Reproductive system | Testicular injury and male infertility |
-
MRI, magnetic resonance imaging; PTSD, post-traumatic stress disorder; CKD, chronic kidney disease.
Epidemiology
COVID-19 has been confirmed in 0.29 billion (290,642,746) individuals worldwide, with over 254,583,687 (88%) recovered cases. By January 3, 2022, 98.2% (3,42,95,407) of COVID-19 patients in India had recovered and had been declared discharged [5]. This group accounts for a sizable portion of the population. Recovery from COVID-19 is a global concern, as growing evidence shows a wide range of sequelae in patients who have returned to microbiological normalization after infection by SARS-CoV-2. Although most patients recover completely within a few weeks, some cases, including those who had mild disease, continue to experience symptoms known as “long hauler” symptoms of COVID-19 or post‐COVID syndrome after their initial recovery [6]. Long-term consequences of COVID-19 are widely documented, although the relative frequency of specific symptoms and diagnoses has yet to be determined [7, 8]. Post-Covid syndrome has been defined by the National Institute for Health and Care Excellence guideline [9], as signs and symptoms that appear during or after an infection consistent with COVID-19, that last for more than 12 weeks, and are not explained by another diagnosis (as shown in Figure 1). Furthermore, the recommendation emphasizes that many persons with post-COVID syndrome suffer from nonspecific symptoms like physical exhaustion, a persistently high fever, and psychological issues. In addition to the aforementioned post COVID syndrome, evidence is mounting indicating COVID sequelae lasting more than three weeks from the onset of first symptoms, which are referred as post-acute COVID-19 [10]. The implications of infection are currently being investigated as this is a newly discovered virus. There is currently no well defined clinical classification for post-COVID-19 symptoms, nor are there any well-defined therapeutic management guidelines. As the numbers of people recovering from COVID-19 are increasing, it is critical to gain a better knowledge of the healthcare difficulties that they are likely to encounter. COVID-19 is now known to be a multi-organ disease with a wide range of symptoms. There are increasing reports of persistent and extended sequelae after acute COVID-19, similar to post-acute viral symptoms observed in survivors of previous virulent coronavirus outbreaks [11].

Definition of long and acute COVID-19.
An observational cohort study conducted in 38 hospitals of United States looked at the outcomes on 1,250 patients that have recovered from COVID-19. 6.7% of patients died over the study period, while 15.1% required re-admission. There were 32.6% of patients who had chronic symptoms, with 18.9% having new or exacerbated symptoms. The most prevalent symptom was dyspnea while walking up the stairs (22.9%), followed by cough (15.4%) and prolonged loss of taste and/or smell (13.1%) [12]. Similar findings have been observed from European studies like in an Italian study, at a mean follow-up of 60 days following the onset of the first symptom, 87.4% of 143 patients discharged from hospital who recovered from acute COVID-19 (referred to as the post-acute COVID-19) reported persistence of symptoms. The most commonly reported symptoms were fatigue (53.1%), dyspnea (43.4%), joint pain (27.3%), and chest pain (21.7%), with 55% of patients continuing to experience three or more symptoms. In this study, 44.1% of patients had a deterioration in quality of life, as judged by the EuroQol visual analogue scale [13]. Another European research study conducted in 150 non-critical COVID-19 survivors in France reported that symptoms persisted in two-thirds of those who were followed for 60 days, with one-third reporting feeling worse than at the outset of acute COVID-19 [14]. Other studies had comparable results, including surveys of 100 COVID-19 survivors in the United Kingdom at 4–8 weeks post-discharge [15] and 120 patients discharged from hospital in France at 100 days after admission [16]. At the time of follow-up, fatigue, dyspnea, and psychological discomfort, such as post-traumatic stress disorder (PTSD), anxiety, depression, and concentration and sleep irregularities, were reported by 30% or more of the study participants. Long-term effects of COVID-19 were assessed in a prospective cohort study from Wuhan, China, using a full in-person evaluation of 1,733 patients at 6 months from onset of symptom (hereby referred to as the post-acute COVID-19 Chinese study) [17]. At least one symptom was recorded by the vast majority of patients (76%). Fatigue/muscular weakness was the most commonly reported symptom (63%), followed by sleep difficulties (26%), and anxiety/depression (23%). Another study in China looked at the effects of COVID-19 on patients three months after hospital discharged. This was a questionnaire-based study that answered questions on five elements of COVID-19 survivors’ symptoms: overall symptoms, respiratory symptoms, cardiovascular symptoms, psychosocial symptoms, and specific symptoms. After three months of discharge, general symptoms were seen in 49% of patients. Among general symptoms, fatigue was seen in 28% of patients. Respiratory symptoms were seen in 39% of patients in which post-activity shortness of breath, and chest distress was seen in 21 and 14% patients, respectively. Cardiovascular symptoms were seen in 13% of patients, and 22.7% of patients reported psychosocial symptoms [18].
These studies provide preliminary data that can be used to identify people who are at high risk of acquiring post-acute COVID-19. The presence or persistence of symptoms (such as dyspnea, fatigue/muscular weakness, and PTSD) in the post-acute COVID-19 setting, as well as a decrease in health-related quality of life scores, pulmonary function abnormalities, and radiographic abnormalities, have all been linked to the severity of illness during acute COVID-19 (measured, for example, by admission to an intensive care unit (ICU) and/or the need for non-invasive and/or invasive mechanical ventilation) [11]. Furthermore, Halpin et al. [15] discovered additional links between pre-existing respiratory disease, higher BMI, older age, and Black, Asian, and minority ethnic (BAME) and dyspnea after 4–8 weeks of follow-up. The post-acute COVID-19 Chinese study demonstrated sex differences, with women more likely to experience tiredness and anxiety/depression at 6 months follow-up, similar to SARS survivors [17]. Other comorbidities are well-known risk factors of increased COVID-19 severity and mortality, such as diabetes, obesity, chronic cardiovascular or kidney disease, cancer, and organ transplantation, but their link with post-acute COVID-19 outcomes in people who have recovered is still unknown. According to one study, matched by propensity score, 10% of COVID-19 cases had one new sequela and 4% had more than one new sequela requiring medical care within a 4-month period commencing 21 days after diagnosis. Compared to the uninfected 2020 group, these rates were higher by 2.9 percent (1 sequela) and 2 percent (>1 sequela); rates were higher by 0.7 percent and 1 percent, respectively, compared to the viral lower respiratory tract illness group. When compared to the 2020 uninfected group, COVID-19 cases had the highest hazard ratios (HR>2.5) for interstitial lung disease, encephalopathy, respiratory failure, pulmonary hypertension, hypercoagulability (deep venous thrombosis, pulmonary embolism), congestive heart failure, arrhythmia, memory disorder, and stroke. HRs peaked in the first month but remained significantly elevated for up to six months, particularly for fatigue, sleep apnea, diabetes, and hypertension, which all increased with age [19, 20]. Although SARS-CoV-2 primarily affects the respiratory system, COVID-19 appears to be a multisystem infection with both overt and subtle health implications, according to many investigations and interim findings [21, 22].
Nonspecific symptoms
There are emerging evidences that a significant number of COVID-19 convalescents experienced weariness weeks after the acute illness has passed. Post-infectious weariness has been found in a variety of viral illnesses months and even years following recovery. Chronic fatigue has been reported as a long-term effect of influenza A(H1N1) virus, SARS-CoV, Ebolavirus, and West Nile virus outbreaks, particularly in those younger than 30 years old. The diagnostic criteria for Chronic Fatigue Syndrome were met in some of these patients. The fundamental cause could be a miscommunication in the inflammatory response pathways, particularly cytokine networks [22]. A COVID-19 infection has been linked to an increased risk of developing a variety of cancers. Following a COVID-19 infection, the mitogen-activated protein kinase and janus kinase/signal transducers and activators of transcription pathways would be activated, resulting in enhanced carcinogenesis, as well as a compromised immune system following a cytokine storm [23].
Otorhinolaryngological sequelae of COVID-19 infection
The olfactory system is thought to act as a viral sensor after infection with SARS-CoV-2, alerting medical personnel to the presence of the disease. A large number of participants reported olfactory and gustatory impairment that did not improve or improved only moderately. The olfactory bulbs are critical in removing invading pathogens from this entry-prone interface early and quickly. Viral clearance is thought to be both rapid and robust, preventing post-mortem detection of virions in patients who have had a long hospital stay and time to autopsy. According to Imam et al. [24], COVID-19-related olfactory dysfunction follows a similar path to post-viral olfactory dysfunction seen in other viral infections such as influenza, rhinovirus, metapneumovirus, or parainfluenza virus. As a result, these adverse effects may become less noticeable over time [24].
Respiratory sequelae of COVID-19 infection
Many patients experience chronic respiratory symptoms weeks to months after being diagnosed with COVID-19. Endothelial and epithelial damage produced by monocyte and neutrophil invasion resulting in acute respiratory distress syndrome (ARDS) is mediated by both viral-dependent and viral-independent processes. The most commonly documented physiologic abnormality in post-acute COVID-19 is a decrease in diffusion capacity, which is directly related to the severity of acute sickness. The persistence of ground-glass opacities is the most common high-resolution lung computed tomography (CT) finding in post-acute COVID-19. Dyspnea, cough, oxygen dependency, difficulties weaning from mechanical ventilation or non-invasive ventilation, fibrotic lung alterations, decreased diffusion capacity, and decreased endurance are common pulmonary sequelae in patients with post-acute COVID-19 syndrome. In post-acute COVID-19, dyspnea is the most common pulmonary symptom (40–50% prevalence at 100 days). Because of shortness of breath, the average 6-min walking distance was significantly lower than the usual reference after a 6-month follow-up. At the 60-day follow-up, about 6% of patients still require supplementary oxygen [12]. At a 30-day follow-up post-discharge, about half of tracheostomy patients were successfully weaned off, according to research from Spain [25]. At a 6-month follow-up, around half of the patients have at least one abnormal CT chest finding (ground-glass opacity, fibrotic alterations).
Pathophysiology
Both viral-dependent (such as SARS-CoV-2 invasion of alveolar epithelial and endothelial cells) and viral-independent (such as immunological damage, including perivascular inflammation) mechanisms contribute to the breakdown of the endothelial–epithelial barrier, with monocyte and neutrophil invasion and extravasation of a protein-rich exudate into the alveolar space, as seen in other forms of ARDS [26]. In the COVID-19 patient`s autopsy report, all phases of diffuse alveolar damage have been described, with organized and localised fibroproliferative diffuse alveolar damage seen later in the disease course, comparable with other ARDS etiologies [27–30]. Cytokines like interleukin-6 (IL-6) and transforming growth factor-β, which have been linked to the development of pulmonary fibrosis and may predispose to bacterial colonization and infection, may trigger this fibrotic condition [31–36]. The histopathologic and single-cell RNA expression patterns in lung tissue were similar to end-stage pulmonary fibrosis without ongoing SARS-CoV-2 infection, suggesting that some people experience accelerated lung fibrosis after the active illness is resolved [37]. COVID-19 patients exhibit pulmonary vascular micro-thrombosis and macro-thrombosis at a rate of 20–30%, which is significantly greater than other critically sick patient populations (1–10%) [11, 37, 38].
Hematologic sequelae
Compared to consumption coagulopathy from disseminated intravascular coagulation (DIC), acute COVID-19 related thromboembolism is secondary to the hyperinflammatory and hypercoagulable state. Thromboembolism is disproportionately high in acute COVID-19 due to hypoxia, endothelial damage, platelet activation, and proinflammatory cytokines. In the post-COVID-19 phase, both the duration and intensity of this hyper-inflammatory state contribute to the risk of thrombotic events [39].
Pathophysiology
COVID-19-associated coagulopathy, unlike consumption coagulopathy associated with disseminated intravascular coagulation, is associated with a hyperinflammatory and hypercoagulable condition [40, 41]. This could explain why acute COVID-19 has a disproportionately high rate of thrombotic rather than bleeding consequences (20–30%) [42]. Endothelial injury, complement activation, platelet activation and platelet–leukocyte interactions, neutrophil extracellular traps, release of pro-inflammatory cytokines, disruption of normal coagulant pathways, and hypoxia are all part of the pathophysiology of thrombo-inflammation, which is similar to the pathophysiology of thrombotic microangiopathy syndromes. The duration and intensity of a hyperinflammatory state are likely associated to the risk of thrombotic problems in the post-acute COVID-19 phase, however, how long this last is still not known [43–49].
Cardiovascular sequelae
Cardiovascular consequences have been documented in asymptomatic COVID-19 patients as well as symptomatic COVID-19 patients. In most persons with Long Covid, the most common symptom is extreme weariness. Other symptoms in patients with cardiac sequelae include chest pain, dyspnea, and palpitations, to name a few. Up to 20–30% of patients with severe COVID-19 who are admitted to the hospital have cardiac involvement, as evidenced by high troponin levels, venous thromboembolism, heart failure, and arrhythmias. Troponin levels beyond a certain threshold have been linked to poor outcomes and greater in-hospital mortality in acutely symptomatic individuals. Increased cardiometabolic demands, myocardial fibrosis or scarring, chronic left ventricular dysfunction, heart failure, arrhythmias, inappropriate sinus tachycardia, and autonomic dysfunctions are all long-term consequences. At the 60 day follow-up, 20% of COVID-19 survivors complained chest pain. At the 60 day follow-up, 10% of COVID-19 survivors experienced palpitations. At six months after receiving COVID-19, 5 and 9% of patients experienced ongoing chest discomfort and palpitations, respectively. In one study, myocardial inflammation observed on cardiac MRI was found in up to 60% of patients more than 2 months following diagnosis. According to retrospective research, the risk of venous thromboembolism in the post-acute COVID-19 context is around 5%. The vast majority of patients with asymptomatic/mild COVID-19, on the other hand, do not have any significant consequences [50].
Pathophysiology
Direct viral invasion, downregulation of ACE-2, inflammation, and the immune response all impact the structural integrity of the myocardial, pericardium, and conduction system, prolonging cardiovascular sequelae in post-acute COVID-19. In 39 cases of COVID-19, autopsy examinations revealed virus in the cardiac tissue of 62.5% of victims [51]. The inflammatory response that follows may result in cardiomyocyte mortality and fibro-fatty displacement of desmosomal proteins that are required for cell-to-cell adhesion [52, 53]. As seen in long-term evaluations of SARS survivors, recovered patients have a continuously elevated cardiometabolic demand [54]. This may be linked to decreased cardiac reserve, corticosteroid use, and renin–angiotensin–aldosterone system dysregulation (RAAS). Re-entrant arrhythmia can be caused by myocardial fibrosis or scarring, as well as cardiomyopathy caused by viral infection [55]. COVID-19 may potentially contribute to arrhythmias by increasing catecholamine levels in response to cytokines such IL-6, IL-1, and tumour necrosis factor-b, which can prolong ventricular action potentials by regulating cardiomyocyte ion channel expression [56]. Adrenergic modulation has previously been linked to autonomic dysfunction during viral infection, resulting in postural orthostatic tachycardia syndrome and inappropriate sinus tachycardia [57]. Endomyocardial biopsy immunohistochemistry revealed significant intramyocardial inflammation with an increase in perforin-positive cells. The number of macrophages, T lymphocytes, and CD45R0 T memory cells have been found to be increased. The amount of cell adhesion molecules (CAM) such as CD 54/ICAM-1 have also seen to be increased [58].
Neuropsychiatric sequelae
The most common symptoms observed in people infected with post acute COVID are headache, anosmia, and myalgia. SARS-CoV-2 infection can damage the central nervous system and cause demyelinating lesions in the spine, which can lead to neuropsychiatric symptoms in the cognitive, emotional, behavioural, and perceptual domains. These neuropsychiatric symptoms, which include cerebrovascular, mental, and neuromuscular diseases, are common in elderly patients and those who have numerous comorbidities or are suffering from a severe illness. During the acute phase, both SARS and MERS are linked to delirium, depression, anxiety, memory loss, and sleeplessness. During the post-illness period, depression, sleeplessness, anxiety, memory loss, and sleep difficulties are common. Dementia, agitation, altered consciousness, and other neuropsychiatric symptoms, such as encephalopathy, encephalitis, depression, anxiety, and post-traumatic stress disorder, affect a large number of COVID-19 patients [59].
Pathophysiology
Direct viral infection, severe systemic inflammation, neuroinflammation, microvascular thrombosis, and neurodegeneration are some of the mechanisms that contribute to neuropathology in COVID-19 [60–62]. SARS-CoV-2 has been found to cause alterations in the brain parenchyma and arteries, potentially as a result of impacts on the blood–brain and blood–cerebrospinal fluid barriers, which drive inflammation in neurons, supporting cells, and the brain vasculature [63–65]. Furthermore, immunological activation levels are linked to cognitive and behavioural changes [66]. Inflammation (chronic low-level brain inflammation), together with a decreased ability to respond to novel antigens and an accumulation of memory T cells (hallmarks of immunosenescence in ageing and tissue injury, respectively) [67], may play a role in COVID-19’s long-term effects. Other theories include faulty lymphatic outflow from circumventricular organs, viral invasion in olfactory epithelial extracellular spaces, and passive diffusion and axonal transport across the olfactory complex. In patients with COVID-19, biomarkers of brain injury, such as higher neurofilament light chain levels in the peripheral blood, have been discovered, along with a more sustained increase in severe infections, implying the likelihood of more chronic neuronal impairment [68–71]. Deconditioning or Post-traumatic stress disorder (PTSD) may play a role in the development of post-COVID brain fog in critically ill COVID-19 patients. COVID-19 brain fog after mild COVID-19, on the other hand, suggests that dysautonomia may play a role. Finally, in the post-critical illness context, long-term cognitive impairment is well established, occurring in 20–40% of patients discharged from an ICU [72, 73].
Renal sequelae
Electrolyte disruption (especially hyperkalaemia) was the most common renal consequence, with a 12.5% incidence, followed by acute kidney injury (AKI) with an 11% incidence. AKI (adjusted hazard ratio (aHR), 1.94; 95% CI, 1.86–2.04), estimated glomerular filtration rate (eGFR) reduction 305 (aHR, 1.25; 955 CI, 1.14–1.37), eGFR decline 40% (aHR, 1.44; 95% CI, 1.37–1.51), eGFR decline 50% (aHR, 1.62; 95% CI, 1.51–1.74), end stage kidney disease (aHR, 2.96; 95% CI, 2.49) to (aHR, 1.66; 95% CI, 1.58–1.74). During hospitalisation, a significant number (20%) of severe COVID-19 patients who required intubation also required renal replacement therapy (RRT). Near discharge, the majority of them did not require dialysis. AKI requiring renal replacement therapy (RRT) occurs in 5% of all hospitalised patients and 20–31% of critically sick patients with acute COVID-19, particularly those who require mechanical ventilation. According to longer-term follow-up studies, patients who require RRT for severe AKI have a high death rate, with a survival probability of 0.46 at 60 days and rates of renal recovery of 84 percent among survivors [74–77].
Pathophysiology
SARS-CoV-2 has been isolated from renal tissue, and the most common finding in cases of COVID-19 renal biopsies and autopsy is acute tubular necrosis. COVID-19-associated nephropathy (COVAN) is a kind of focal segmental glomerulosclerosis that includes involution of the glomerular tuft as well as acute tubular damage. It is considered to occur in response to interferon and chemokine activation. SARS-association CoV-2’s with apolipoprotein L1 risk alleles suggests that, like the human immunodeficiency virus and other viruses, it functions as a second hit in vulnerable patients. Renal microcirculation thrombosis may possibly play a role in the development of renal damage [78–85].
Endocrine sequelae
Post-acute COVID-19 endocrine symptoms are caused by viral infection, inflammatory, and immunologic damage. Weeks after resolving acute COVID-19 symptoms, isolated case reports of diabetic ketoacidosis (DKA), subacute, and Hashimoto thyroiditis or Graves’ disease have been reported. Immobilization, steroid use, and vitamin D insufficiency during COVID-19 healing may all play a role in bone demineralization [86, 87].
Pathophysiology
In the post-acute COVID-19 scenario, endocrine symptoms could be the result of direct viral damage, immunological and inflammatory injury, as well as iatrogenic sequelae. Even if initially linked with DKA, pre-existing diabetes may become obvious during the acute phase of COVID-19 and may normally be treated long term with medicines other than insulin. There is no evidence that pancreatic cells are permanently damaged. Although some studies have found ACE2 and transmembrane serine protease (TMPRSS2; the protease involved in SARS-CoV-2 cell entry) expression in cells, the primary insulin production deficit is most likely mediated by inflammation or the infection stress response, as well as peripheral insulin resistance. There is currently no indication that COVID-19-related diabetes can be reversed after the acute phase, nor that the outcomes differ in COVID-19 long haulers. COVID-19 also shows that systemic inflammation, immobility, corticosteroid exposure, vitamin D deficiency, and cessation of antiresorptive or anabolic medications for osteoporosis are all risk factors for bone demineralization [88–90].
Gastrointestinal and hepatobiliary sequelae
In COVID-19 survivors, no significant gastrointestinal or hepatobiliary effects have been documented. COVID-19 exhibits prolonged viral faecal shedding, with viral ribonucleic acid detectable for an average of 28 days after the onset of SARS-CoV-2 infection symptoms and for an average of 11 days following negative respiratory samples [91–93]. COVID-19 has the potential to change the gut microbiota, favouring opportunistic infectious microbes while reducing beneficial commensals [94, 95]. The gut microbiota’s ability to influence the course of respiratory infections (gut–lung axis) has previously been observed in influenza and other respiratory diseases. Faecalibacterium prausnitzii, a butyrate-producing anaerobe associated with excellent health, was found to be inversely related to disease severity in COVID-19. COVID-19’s long-term effects on the gastrointestinal system, particularly post-infectious irritable bowel syndrome and dyspepsia, are now being studied [96–98].
Dermatologic sequelae
In an international investigation of 716 people with COVID-19, dermatologic manifestations of COVID-19 appeared after (64%) or concurrently with (15%) other acute COVID-19 symptoms, with an average latency of 7.9 days in adults from the time of upper respiratory symptoms to dermatologic findings. In the post-acute COVID-19 Chinese trial, only 3% of patients had a skin rash after 6 months. Hair loss was the most common dermatologic symptom, affecting roughly 20% of patients. Hair loss may be caused by telogen effluvium, which is caused by a viral infection or a stress response. Ongoing research could shed light on illness pathways involving the immune system or inflammation [99–101].
Multisystem inflammatory syndrome in children (MIS-C)
MIS-C is a new clinical entity defined by fever, multiorgan dysfunction, and elevated inflammatory markers in people under the age of 21 years who have recently or currently been infected with SARS-CoV-2. Complement activation, the production of autoantibodies (viral host mimicry), and excessive cytokines from T-cell stimulation are the most likely underlying mechanisms [102].
Pathology and pathophysiology
The emergence of MIS-C (which occurred about a month after peak COVID-19) and the fact that most patients are negative for acute infection but positive for antibodies suggest that MIS-C is the result of an abnormal acquired immune response rather than acute viral infection. Kawasaki disease and toxic shock syndrome may provide insights into the pathophysiology of MIS-C, with possible injury mechanisms related to immune complexes, complement activation, autoantibody formation through viral host mimicry, and massive cytokine release related to superantigen stimulation of T cells [102–104].
Reproductive system
Because the human testis expresses ACE2, an infection could result in potential testicular injury and male infertility [105, 106].
Nutrition and rehabilitation considerations
Like other critical illnesses, severe COVID-19 illness induces catabolic muscle loss, feeding difficulties, and fragility, all of which are linked to a higher risk of poor outcome. Malnutrition was found in 26–45% of COVID-19 patients in an Italian study using the Malnutrition Universal Screening Tool. Nutritional support protocols for patients (many of whom had respiratory discomfort, nausea, diarrhoea, and anorexia, resulting in a drop in food intake) are still being developed. All post-acute COVID-19 follow-up studies that included assessments of health-related quality of life and functional capacity measures, including the post-acute COVID-19 Chinese research, found severe deficits in these categories. Early rehabilitation therapies are being examined in ongoing clinical research due to the severity of the systemic inflammatory response associated with severe COVID-19 and the resulting weakness. They’ve been shown to be both safe and efficacious in critically ill patients with ARDS, as well as in preliminary COVID-19 investigations. Acute COVID-19 survivors require frequent assessment of swallowing function, nutritional condition, and functional independence in model COVID-19 rehabilitation units like those in Italy [11, 107–111].
Conclusion
Despite the fact that most immediate COVID-19 consequences in young, previously healthy adults are temporary, there are signs of long-term multi-organ involvement. The huge global burden of cases suggests that COVID-19 sequelae will most likely be a problem. Residual deficits in multi-organ function as well as mental health and neurological sequelae, such as post-viral fatigue syndrome, should be closely monitored. Further research to detect long-term structural and functional damage is need of the hour. Patients with post-viral fatigue syndrome and mental health issues could be followed up on a regular basis using questionnaires to track their progress. In general, follow-up study test batteries should be carefully constructed to detect subtle, long-term effects. Given the pandemic’s global scope, it’s clear that the healthcare demands of people suffering from COVID-19 complications will continue to rise for the foreseeable future. To meet this issue, current outpatient infrastructure will be leveraged, scalable healthcare models will be developed, and cross-disciplinary collaboration will be required for long-term mental and physical health of COVID-19 survivors.
-
Research funding: None.
-
Author contribution: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Competing interests: Yes, all authors state that there is no conflict of interest.
-
Informed consent: Not required.
-
Ethical approval: No requirement of ethical approval.
References
1. World health organization (WHO). [Homepage in Internet] Director-General’s opening remarks at the media briefing on COVID-19; 2020. Available from: https://www.who.int/dg/speeches/detail/who-director-general-s-opening-remarks-at-the-media-briefing-on-covid-19---11-march-2020. [Accessed 25 Sep 2020].Search in Google Scholar
2. Bansal, M. Cardiovascular disease and COVID-19. Diabetes Metabol Syndr 2020;14:247–50. https://doi.org/10.1016/j.dsx.2020.03.013.Search in Google Scholar PubMed PubMed Central
3. Zheng, J. SARS-CoV-2: an emerging coronavirus that causes a global threat. Int J Biol Sci 2020;16:1678–85. https://doi.org/10.7150/ijbs.45053.Search in Google Scholar PubMed PubMed Central
4. Hamming, I, Timens, W, Bulthuis, ML, Lely, AT, Navis, G, van Goor, H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol 2004;203:631–7. https://doi.org/10.1002/path.1570.Search in Google Scholar PubMed PubMed Central
5. MoHFW [Home page on Internet]. Available from: https://www.mohfw.gov.in/. [Accessed 4 Jan 2021].Search in Google Scholar
6. Jiang, DH, McCoy, RG. Planning for the post-COVID syndrome: how payers can mitigate long-term complications of the pandemic. J Gen Intern Med 2020;35:3036–9. https://doi.org/10.1007/s11606-020-06042-3.Search in Google Scholar PubMed PubMed Central
7. Borg, K, Stam, HJ. Rehabilitation of post-Covid - 19 syndrome - once again a call for action. J Rehabil Med 2021;53:jrm00132. https://doi.org/10.2340/16501977-2783.Search in Google Scholar PubMed PubMed Central
8. Rubin, R. As their numbers grow, COVID-19 “long haulers” stump experts. JAMA 2020;324:1381–3. https://doi.org/10.1001/jama.2020.17709.Search in Google Scholar PubMed
9. The National Institute for Health and Care Excellence. [Home page in Internet] NICE, SIGN and RCGP set out further details about the UK guideline on management of the long-term effects of COVID-19. Available from: https://www.nice.org.uk/. [Accessed 30 Oct 2021].Search in Google Scholar
10. Greenhalgh, T, Knight, M, Buxton, M, Husain, L. Management of post-acute Covid-19 in primary care. BMJ 2020;370:m3026. https://doi.org/10.1136/bmj.m3026.Search in Google Scholar PubMed
11. Nalbandian, A, Sehgal, K, Gupta, A, Madhavan, MV, McGroder, C, Stevens, JS, et al.. Post-acute COVID-19 syndrome. Nat Med 2021;27:601–15. https://doi.org/10.1038/s41591-021-01283-z.Search in Google Scholar PubMed PubMed Central
12. Chopra, V, Flanders, SA, O’Malley, M, Malani, AN, Prescott, HC. Sixty-day outcomes among patients hospitalized with COVID-19. Ann Intern Med 2021;174:576–8. https://doi.org/10.7326/m20-5661.Search in Google Scholar
13. Carfì, A, Bernabei, R, Landi, F. Gemelli against COVID-19 post-acute care study group. Persistent symptoms in patients after acute COVID-19. JAMA 2020;324:603–5. https://doi.org/10.1001/jama.2020.12603.Search in Google Scholar PubMed PubMed Central
14. Carvalho-Schneider, C, Laurent, E, Lemaignen, A, Beaufils, E, Bourbao-Tournois, C, Laribi, S, et al.. Follow-up of adults with noncritical COVID-19 two months after symptom onset. Clin Microbiol Infect 2021;27:258–63. https://doi.org/10.1016/j.cmi.2020.09.052.Search in Google Scholar PubMed PubMed Central
15. Halpin, SJ, McIvor, C, Whyatt, G, Adams, A, Harvey, O, McLean, L, et al.. Postdischarge symptoms and rehabilitation needs in survivors of COVID-19 infection: a cross-sectional evaluation. J Med Virol 2021;93:1013–22. https://doi.org/10.1002/jmv.26368.Search in Google Scholar PubMed
16. Garrigues, E, Janvier, P, Kherabi, Y, Le Bot, A, Hamon, A, Gouze, H, et al.. Post-discharge persistent symptoms and health-related quality of life after hospitalization for COVID-19. J Infect 2020;81:e4–6. https://doi.org/10.1016/j.jinf.2020.08.029.Search in Google Scholar PubMed PubMed Central
17. Huang, C, Huang, L, Wang, Y, Li, X, Ren, L, Gu, X, et al.. 6-month consequences of COVID-19 in patients discharged from hospital: a cohort study. Lancet 2021;397:220–32. https://doi.org/10.1016/s0140-6736(20)32656-8.Search in Google Scholar PubMed PubMed Central
18. Xiong, Q, Xu, M, Li, J, Liu, Y, Zhang, J, Xu, Y, et al.. Clinical sequelae of COVID-19 survivors in Wuhan, China: a single-centre longitudinal study. Clin Microbiol Infect 2021;27:89–95. https://doi.org/10.1016/j.cmi.2020.09.023.Search in Google Scholar PubMed PubMed Central
19. Williamson, EJ, Walker, AJ, Bhaskaran, K, Bacon, S, Bates, C, Morton, CE, et al.. Factors associated with COVID-19-related death using OpenSAFELY. Nature 2020;584:430–6. https://doi.org/10.1038/s41586-020-2521-4.Search in Google Scholar PubMed PubMed Central
20. Gupta, A, Madhavan, MV, Sehgal, K, Nair, N, Mahajan, S, Sehrawat, TS, et al.. Extrapulmonary manifestations of COVID-19. Nat Med 2020;26:1017–32. https://doi.org/10.1038/s41591-020-0968-3.Search in Google Scholar PubMed
21. Daugherty, SE, Guo, Y, Heath, K, Dasmariñas, MC, Jubilo, KG, Samranvedhya, J, et al.. Risk of clinical sequelae after the acute phase of SARS-CoV-2 infection: retrospective cohort study. BMJ 2021;373:n1098. https://doi.org/10.1136/bmj.n1098.Search in Google Scholar PubMed PubMed Central
22. Islam, MF, Cotler, J, Jason, LA. Post-viral fatigue and COVID-19: lessons from past epidemics. Fatigue: Biomed Health Behav 2020;8:61–9. https://doi.org/10.1080/21641846.2020.1778227.Search in Google Scholar
23. Hays, P. Clinical sequelae of the novel coronavirus: does COVID-19 infection predispose patients to cancer?. Future Oncol 2020;16:1463–74. https://doi.org/10.2217/fon-2020-0300.Search in Google Scholar PubMed PubMed Central
24. Imam, SA, Lao, WP, Reddy, P, Nguyen, SA, Schlosser, RJ. Is SARS-CoV-2 (COVID-19) postviral olfactory dysfunction (PVOD) different from other PVOD?. World J Otorhinolaryngol Head Neck Surg 2020;6(1 Suppl):S26–32. https://doi.org/10.1016/j.wjorl.2020.05.004.Search in Google Scholar PubMed PubMed Central
25. Martin-Villares, C, Perez Molina-Ramirez, C, Bartolome-Benito, M, Bernal-Sprekelsen, M, COVID ORL ESP Collaborative Group (*). Outcome of 1890 tracheostomies for critical COVID-19 patients: a national cohort study in Spain. Eur Arch Oto-Rhino-Laryngol 2021;278:1605–12.10.1007/s00405-020-06220-3Search in Google Scholar PubMed PubMed Central
26. Huppert, LA, Matthay, MA, Ware, LB. Pathogenesis of acute respiratory distress syndrome. Semin Respir Crit Care Med 2019;40:31–9. https://doi.org/10.1055/s-0039-1683996.Search in Google Scholar PubMed PubMed Central
27. Carsana, L, Sonzogni, A, Nasr, A, Rossi, RS, Pellegrinelli, A, Zerbi, P, et al.. Pulmonary post-mortem findings in a series of COVID-19 cases from northern Italy: a two-centre descriptive study. Lancet Infect Dis 2020;20:1135–40. https://doi.org/10.1016/s1473-3099(20)30434-5.Search in Google Scholar PubMed PubMed Central
28. Schaller, T, Hirschbühl, K, Burkhardt, K, Braun, G, Trepel, M, Märkl, B, et al.. Postmortem examination of patients with COVID-19. JAMA 2020;323:2518–20. https://doi.org/10.1001/jama.2020.8907.Search in Google Scholar PubMed PubMed Central
29. Burnham, EL, Janssen, WJ, Riches, DW, Moss, M, Downey, GP. The fibroproliferative response in acute respiratory distress syndrome: mechanisms and clinical significance. Eur Respir J 2014;43:276–85. https://doi.org/10.1183/09031936.00196412.Search in Google Scholar PubMed PubMed Central
30. De Michele, S, Sun, Y, Yilmaz, MM, Katsyv, I, Salvatore, M, Dzierba, AL, et al.. Forty postmortem examinations in COVID-19 patients. Am J Clin Pathol 2020;154:748–60. https://doi.org/10.1093/ajcp/aqaa156.Search in Google Scholar PubMed PubMed Central
31. Coker, RK, Laurent, GJ, Jeffery, PK, du Bois, RM, Black, CM, McAnulty, RJ. Localisation of transforming growth factor beta1 and beta3 mRNA transcripts in normal and fibrotic human lung. Thorax 2001;56:549–56. https://doi.org/10.1136/thorax.56.7.549.Search in Google Scholar PubMed PubMed Central
32. Le, TT, Karmouty-Quintana, H, Melicoff, E, Le, TT, Weng, T, Chen, NY, et al.. Blockade of IL-6 Trans signaling attenuates pulmonary fibrosis. J Immunol 2014;193:3755–68. https://doi.org/10.4049/jimmunol.1302470.Search in Google Scholar PubMed PubMed Central
33. Moodley, YP, Scaffidi, AK, Misso, NL, Keerthisingam, C, McAnulty, RJ, Laurent, GJ, et al.. Fibroblasts isolated from normal lungs and those with idiopathic pulmonary fibrosis differ in interleukin-6/gp130-mediated cell signaling and proliferation. Am J Pathol 2003;163:345–54. https://doi.org/10.1016/s0002-9440(10)63658-9.Search in Google Scholar PubMed PubMed Central
34. Chen, G, Wu, D, Guo, W, Cao, Y, Huang, D, Wang, H, et al.. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 2020;130:2620–9. https://doi.org/10.1172/jci137244.Search in Google Scholar PubMed PubMed Central
35. Hendaus, MA, Jomha, FA. Covid-19 induced superimposed bacterial infection. J Biomol Struct Dyn 2021;39:4185–91. https://doi.org/10.1080/07391102.2020.1772110.Search in Google Scholar PubMed
36. Hendaus, MA, Jomha, FA, Alhammadi, AH. Virus-induced secondary bacterial infection: a concise review. Therapeut Clin Risk Manag 2015;11:1265–71. https://doi.org/10.2147/tcrm.s87789.Search in Google Scholar
37. Ackermann, M, Verleden, SE, Kuehnel, M, Haverich, A, Welte, T, Laenger, F, et al.. Pulmonary Vascular endothelialitis, thrombosis, and angiogenesis in COVID-19. N Engl J Med 2020;383:120–8. https://doi.org/10.1056/nejmoa2015432.Search in Google Scholar PubMed PubMed Central
38. Bharat, A, Querrey, M, Markov, NS, Kim, S, Kurihara, C, Garza-Castillon, R, et al.. Lung transplantation for patients with severe COVID-19. Sci Transl Med 2020;12:eabe4282. https://doi.org/10.1126/scitranslmed.abe4282.Search in Google Scholar PubMed PubMed Central
39. Chaudhary, R, Kreutz, RP, Bliden, KP, Tantry, US, Gurbel, PA. Personalizing antithrombotic therapy in COVID-19: role of thromboelastography and thromboelastometry. Thromb Haemostasis 2020;120:1594–6. https://doi.org/10.1055/s-0040-1714217.Search in Google Scholar PubMed
40. Pavoni, V, Gianesello, L, Pazzi, M, Stera, C, Meconi, T, Frigieri, FC. Evaluation of coagulation function by rotation thromboelastometry in critically ill patients with severe COVID-19 pneumonia. J Thromb Thrombolysis 2020;50:281–6. https://doi.org/10.1007/s11239-020-02130-7.Search in Google Scholar PubMed PubMed Central
41. Connors, JM, Levy, JH. COVID-19 and its implications for thrombosis and anticoagulation. Blood 2020;135:2033–40. https://doi.org/10.1182/blood.2020006000.Search in Google Scholar PubMed PubMed Central
42. Varga, Z, Flammer, AJ, Steiger, P, Haberecker, M, Andermatt, R, Zinkernagel, AS, et al.. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020;395:1417–8. https://doi.org/10.1016/s0140-6736(20)30937-5.Search in Google Scholar
43. Ramlall, V, Thangaraj, PM, Meydan, C, Foox, J, Butler, D, Kim, J, et al.. Immune complement and coagulation dysfunction in adverse outcomes of SARS-CoV-2 infection. Nat Med 2020;26:1609–15. https://doi.org/10.1038/s41591-020-1021-2.Search in Google Scholar PubMed PubMed Central
44. Skendros, P, Mitsios, A, Chrysanthopoulou, A, Mastellos, DC, Metallidis, S, Rafailidis, P, et al.. Complement and tissue factor-enriched neutrophil extracellular traps are key drivers in COVID-19 immunothrombosis. J Clin Invest 2020;130:6151–7. https://doi.org/10.1172/jci141374.Search in Google Scholar PubMed PubMed Central
45. Hottz, ED, Azevedo-Quintanilha, IG, Palhinha, L, Teixeira, L, Barreto, EA, Pão, CRR, et al.. Platelet activation and platelet-monocyte aggregate formation trigger tissue factor expression in patients with severe COVID-19. Blood 2020;136:1330–41. https://doi.org/10.1182/blood.2020007252.Search in Google Scholar PubMed PubMed Central
46. Barrett, TJ, Lee, AH, Xia, Y, Lin, LH, Black, M, Cotzia, P, et al.. Platelet and vascular biomarkers associate with thrombosis and death in coronavirus disease. Circ Res 2020;127:945–7. https://doi.org/10.1161/circresaha.120.317803.Search in Google Scholar PubMed PubMed Central
47. Middleton, EA, He, XY, Denorme, F, Campbell, RA, Ng, D, Salvatore, SP, et al.. Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood 2020;136:1169–79. https://doi.org/10.1182/blood.2020007008.Search in Google Scholar PubMed PubMed Central
48. Nougier, C, Benoit, R, Simon, M, Desmurs-Clavel, H, Marcotte, G, Argaud, L, et al.. Hypofibrinolytic state and high thrombin generation may play a major role in SARS-COV2 associated thrombosis. J Thromb Haemostasis 2020;18:2215–9. https://doi.org/10.1111/jth.15016.Search in Google Scholar PubMed PubMed Central
49. Merrill, JT, Erkan, D, Winakur, J, James, JA. Emerging evidence of a COVID-19 thrombotic syndrome has treatment implications. Nat Rev Rheumatol 2020;16:581–9. https://doi.org/10.1038/s41584-020-0474-5.Search in Google Scholar PubMed PubMed Central
50. Directorate General of Health Sciences Ministry of Health and Family Welfare Government of India. [Home page in internet] National comprehensive guidelines for management of post-Covid cardiovascular sequelae. [Available from: https://dghs.gov.in/WriteReadData/News/2021100103542077256241CARDIOLOGY_MODULE.pdf. [Accessed 15 Jan 2021].Search in Google Scholar
51. Lindner, D, Fitzek, A, Bräuninger, H, Aleshcheva, G, Edler, C, Meissner, K, et al.. Association of cardiac infection with SARS-CoV-2 in confirmed COVID-19 autopsy cases. JAMA Cardiol 2020;5:1281–5. https://doi.org/10.1001/jamacardio.2020.3551.Search in Google Scholar PubMed PubMed Central
52. Gemayel, C, Pelliccia, A, Thompson, PD. Arrhythmogenic right ventricular cardiomyopathy. J Am Coll Cardiol 2001;38:1773–81. https://doi.org/10.1016/s0735-1097(01)01654-0.Search in Google Scholar PubMed
53. Siripanthong, B, Nazarian, S, Muser, D, Deo, R, Santangeli, P, Khanji, MY, et al.. Recognizing COVID-19-related myocarditis: the possible pathophysiology and proposed guideline for diagnosis and management. Heart Rhythm 2020;17:1463–71. https://doi.org/10.1016/j.hrthm.2020.05.001.Search in Google Scholar PubMed PubMed Central
54. Wu, Q, Zhou, L, Sun, X, Yan, Z, Hu, C, Wu, J, et al.. Altered lipid metabolism in recovered SARS patients twelve years after infection. Sci Rep 2017;7:9110. https://doi.org/10.1038/s41598-017-09536-z.Search in Google Scholar PubMed PubMed Central
55. Liu, PP, Blet, A, Smyth, D, Li, H. The science underlying COVID-19: implications for the cardiovascular system. Circulation 2020;142:68–78. https://doi.org/10.1161/circulationaha.120.047549.Search in Google Scholar PubMed
56. Lazzerini, PE, Laghi-Pasini, F, Boutjdir, M, Capecchi, PL. Cardioimmunology of arrhythmias: the role of autoimmune and inflammatory cardiac channelopathies. Nat Rev Immunol 2019;19:63–4. https://doi.org/10.1038/s41577-018-0098-z.Search in Google Scholar PubMed
57. Agarwal, AK, Garg, R, Ritch, A, Sarkar, P. Postural orthostatic tachycardia syndrome. Postgrad Med 2007;83:478–80. https://doi.org/10.1136/pgmj.2006.055046.Search in Google Scholar PubMed PubMed Central
58. Escher, F, Pietsch, H, Aleshcheva, G, Bock, T, Baumeier, C, Elsaesser, A, et al.. Detection of viral SARS-CoV-2 genomes and histopathological changes in endomyocardial biopsies. ESC Heart Fail 2020;7:2440–7. https://doi.org/10.1002/ehf2.12805.Search in Google Scholar PubMed PubMed Central
59. Han, Y, Yuan, K, Wang, Z, Liu, WJ, Lu, ZA, Liu, L, et al.. Neuropsychiatric manifestations of COVID-19, potential neurotropic mechanisms, and therapeutic interventions. Transl Psychiatry 2021;11:499. https://doi.org/10.1038/s41398-021-01629-8.Search in Google Scholar PubMed PubMed Central
60. Pilotto, A, Padovani, A, ENCOVID-BIO Network. Reply to the letter “COVID-19-Associated encephalopathy and cytokine-mediated neuroinflammation”. Ann Neurol 2020;88:861–2. https://doi.org/10.1002/ana.25856.Search in Google Scholar PubMed PubMed Central
61. South, K, McCulloch, L, McColl, BW, Elkind, MS, Allan, SM, Smith, CJ. Preceding infection and risk of stroke: an old concept revived by the COVID-19 pandemic. Int J Stroke 2020;15:722–32. https://doi.org/10.1177/1747493020943815.Search in Google Scholar PubMed PubMed Central
62. Desforges, M, Le Coupanec, A, Stodola, JK, Meessen-Pinard, M, Talbot, PJ. Human coronaviruses: viral and cellular factors involved in neuroinvasiveness and neuropathogenesis. Virus Res 2014;194:145–58. https://doi.org/10.1016/j.virusres.2014.09.011.Search in Google Scholar PubMed PubMed Central
63. Romero-Sánchez, CM, Díaz-Maroto, I, Fernández-Díaz, E, Sánchez-Larsen Á, Layos-Romero, A, García-García, J, et al.. Neurologic manifestations in hospitalized patients with COVID-19: the ALBACOVID registry. Neurology 2020;95:e1060–70.10.1212/WNL.0000000000009937Search in Google Scholar PubMed PubMed Central
64. Reichard, RR, Kashani, KB, Boire, NA, Constantopoulos, E, Guo, Y, Lucchinetti, CF. Neuropathology of COVID-19: a spectrum of vascular and acute disseminated encephalomyelitis (ADEM)-like pathology. Acta Neuropathol 2020;140:1–6. https://doi.org/10.1007/s00401-020-02166-2.Search in Google Scholar PubMed PubMed Central
65. Bortolato, B, Carvalho, AF, Soczynska, JK, Perini, GI, McIntyre, RS. The involvement of TNF-α in cognitive dysfunction associated with major depressive disorder: an opportunity for domain specific treatments. Curr Neuropharmacol 2015;13:558–76. https://doi.org/10.2174/1570159x13666150630171433.Search in Google Scholar PubMed PubMed Central
66. Aiello, A, Farzaneh, F, Candore, G, Caruso, C, Davinelli, S, Gambino, CM, et al.. Immunosenescence and its hallmarks: how to oppose aging strategically? A review of potential options for therapeutic intervention. Front Immunol 2019;10:2247. https://doi.org/10.3389/fimmu.2019.02247.Search in Google Scholar PubMed PubMed Central
67. Perrin, R, Riste, L, Hann, M, Walther, A, Mukherjee, A, Heald, A. Into the looking glass: post-viral syndrome post COVID-19. Med Hypotheses 2020;144:110055. https://doi.org/10.1016/j.mehy.2020.110055.Search in Google Scholar PubMed PubMed Central
68. Morbini, P, Benazzo, M, Verga, L, Pagella, FG, Mojoli, F, Bruno, R, et al.. Ultrastructural evidence of direct viral damage to the olfactory complex in patients testing positive for SARS-CoV-2. JAMA Otolaryngol Head Neck Surg 2020;146:972–3. https://doi.org/10.1001/jamaoto.2020.2366.Search in Google Scholar PubMed
69. Ameres, M, Brandstetter, S, Toncheva, AA, Kabesch, M, Leppert, D, Kuhle, J, et al.. Association of neuronal injury blood marker neurofilament light chain with mild-to-moderate COVID-19. J Neurol 2020;267:3476–8. https://doi.org/10.1007/s00415-020-10050-y.Search in Google Scholar PubMed PubMed Central
70. Kanberg, N, Ashton, NJ, Andersson, LM, Yilmaz, A, Lindh, M, Nilsson, S, et al.. Neurochemical evidence of astrocytic and neuronal injury commonly found in COVID-19. Neurology 2020;95:e1754–59. https://doi.org/10.1212/wnl.0000000000010111.Search in Google Scholar
71. Novak, P. Post COVID-19 syndrome associated with orthostatic cerebral hypoperfusion syndrome, small fiber neuropathy and benefit of immunotherapy: a case report. eNeurologicalSci 2020;21:100276. https://doi.org/10.1016/j.ensci.2020.100276.Search in Google Scholar PubMed PubMed Central
72. Miglis, MG, Goodman, BP, Chémali, KR, Stiles, L. Re: ’Post-COVID-19 chronic symptoms’ by Davido et al. Clin Microbiol Infect 2021;27:494. https://doi.org/10.1016/j.cmi.2020.08.028.Search in Google Scholar PubMed PubMed Central
73. Sakusic, A, Rabinstein, AA. Cognitive outcomes after critical illness. Curr Opin Crit Care 2018;24:410–4. https://doi.org/10.1097/mcc.0000000000000527.Search in Google Scholar
74. Kunutsor, SK, Laukkanen, JA. Renal complications in COVID-19: a systematic review and meta-analysis. Ann Med 2020;52:345–53. https://doi.org/10.1080/07853890.2020.1790643.Search in Google Scholar PubMed PubMed Central
75. Bowe, B, Xie, Y, Xu, E, Al-Aly, Z. Kidney outcomes in long COVID. J Am Soc Nephrol 2021;32:2851–62. https://doi.org/10.1681/asn.2021060734.Search in Google Scholar
76. Gupta, S, Hayek, SS, Wang, W, Chan, L, Mathews, KS, Melamed, ML, et al.. STOP-COVID investigators. Factors associated with death in critically ill patients with coronavirus disease 2019 in the US. JAMA Intern Med 2020;180:1436–47. https://doi.org/10.1001/jamainternmed.2020.3596.Search in Google Scholar PubMed PubMed Central
77. Stevens, JS, King, KL, Robbins-Juarez, SY, Khairallah, P, Toma, K, Alvarado Verduzco, H, et al.. High rate of renal recovery in survivors of COVID-19 associated acute renal failure requiring renal replacement therapy. PLoS One 2020;15:e0244131. https://doi.org/10.1371/journal.pone.0244131.Search in Google Scholar PubMed PubMed Central
78. Su, H, Yang, M, Wan, C, Yi, LX, Tang, F, Zhu, HY, et al.. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int 2020;98:219–27. https://doi.org/10.1016/j.kint.2020.04.003.Search in Google Scholar PubMed PubMed Central
79. Kudose, S, Batal, I, Santoriello, D, Xu, K, Barasch, J, Peleg, Y, et al.. Kidney biopsy findings in patients with COVID-19. J Am Soc Nephrol 2020;31:1959–68. https://doi.org/10.1681/asn.2020060802.Search in Google Scholar PubMed PubMed Central
80. Sharma, P, Uppal, NN, Wanchoo, R, Shah, HH, Yang, Y, Parikh, R, et al.. Northwell nephrology COVID-19 research consortium. COVID-19-associated kidney injury: a case series of kidney biopsy findings. J Am Soc Nephrol 2020;31:1948–58. https://doi.org/10.1681/asn.2020050699.Search in Google Scholar
81. Golmai, P, Larsen, CP, DeVita, MV, Wahl, SJ, Weins, A, Rennke, HG, et al.. Histopathologic and ultrastructural findings in postmortem kidney biopsy material in 12 patients with AKI and COVID-19. J Am Soc Nephrol 2020;31:1944–7. https://doi.org/10.1681/asn.2020050683.Search in Google Scholar PubMed PubMed Central
82. Santoriello, D, Khairallah, P, Bomback, AS, Xu, K, Kudose, S, Batal, I, et al.. Postmortem kidney pathology findings in patients with COVID-19. J Am Soc Nephrol 2020;31:2158–67. https://doi.org/10.1681/asn.2020050744.Search in Google Scholar
83. Velez, JCQ, Caza, T, Larsen, CP. COVAN is the new HIVAN: the re-emergence of collapsing glomerulopathy with COVID-19. Nat Rev Nephrol 2020;16:565–7. https://doi.org/10.1038/s41581-020-0332-3.Search in Google Scholar PubMed PubMed Central
84. Peleg, Y, Kudose, S, D’Agati, V, Siddall, E, Ahmad, S, Nickolas, T, et al.. Acute kidney injury due to collapsing glomerulopathy following COVID-19 infection. Kidney Int Rep 2020;5:940–5. https://doi.org/10.1016/j.ekir.2020.04.017.Search in Google Scholar PubMed PubMed Central
85. Jhaveri, KD, Meir, LR, Flores Chang, BS, Parikh, R, Wanchoo, R, Barilla-LaBarca, ML, et al.. Thrombotic microangiopathy in a patient with COVID-19. Kidney Int 2020;98:509–12. https://doi.org/10.1016/j.kint.2020.05.025.Search in Google Scholar PubMed PubMed Central
86. Brancatella, A, Ricci, D, Viola, N, Sgrò, D, Santini, F, Latrofa, F. Subacute thyroiditis after sars-CoV-2 infection. J Clin Endocrinol Metab 2020;105:dgaa276. https://doi.org/10.1210/clinem/dgaa276.Search in Google Scholar PubMed PubMed Central
87. Rubino, F, Amiel, SA, Zimmet, P, Alberti, G, Bornstein, S, Eckel, RH, et al.. New-onset diabetes in covid-19. N Engl J Med 2020;383:789–90. https://doi.org/10.1056/nejmc2018688.Search in Google Scholar
88. Gentile, S, Strollo, F, Mambro, A, Ceriello, A. COVID-19, ketoacidosis and new-onset diabetes: are there possible cause and effect relationships among them? Diabetes Obes Metabol 2020;22:2507–8. https://doi.org/10.1111/dom.14170.Search in Google Scholar PubMed PubMed Central
89. Yang, JK, Lin, SS, Ji, XJ, Guo, LM. Binding of SARS coronavirus to its receptor damages islets and causes acute diabetes. Acta Diabetol 2010;47:193–9. https://doi.org/10.1007/s00592-009-0109-4.Search in Google Scholar PubMed PubMed Central
90. Salvio, G, Gianfelice, C, Firmani, F, Lunetti, S, Balercia, G, Giacchetti, G. Bone metabolism in SARS-CoV-2 disease: possible osteoimmunology and gender implications. Clin Rev Bone Miner Metabol 2020;1–7. https://doi.org/10.1007/s12018-020-09274-3. 32904892.Search in Google Scholar PubMed PubMed Central
91. Cheung, KS, Hung, IFN, Chan, PPY, Lung, KC, Tso, E, Liu, R, et al.. Gastrointestinal manifestations of SARS-CoV-2 infection and virus load in fecal samples from a Hong Kong cohort: systematic review and meta-analysis. Gastroenterology 2020;159:81–95. https://doi.org/10.1053/j.gastro.2020.03.065.Search in Google Scholar PubMed PubMed Central
92. Wu, Y, Guo, C, Tang, L, Hong, Z, Zhou, J, Dong, X, et al.. Prolonged presence of SARS-CoV-2 viral RNA in faecal samples. Lancet Gastroenterol Hepatol 2020;5:434–5. https://doi.org/10.1016/s2468-1253(20)30083-2.Search in Google Scholar PubMed PubMed Central
93. Xiao, F, Tang, M, Zheng, X, Liu, Y, Li, X, Shan, H. Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterology 2020;158:1831–3. https://doi.org/10.1053/j.gastro.2020.02.055.Search in Google Scholar PubMed PubMed Central
94. Xu, Y, Li, X, Zhu, B, Liang, H, Fang, C, Gong, Y, et al.. Characteristics of pediatric SARS-CoV-2 infection and potential evidence for persistent fecal viral shedding. Nat Med 2020;26:502–5. https://doi.org/10.1038/s41591-020-0817-4.Search in Google Scholar PubMed PubMed Central
95. Zuo, T, Zhang, F, Lui, GCY, Yeoh, YK, Li, AYL, Zhan, H, et al.. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology 2020;159:944–55. https://doi.org/10.1053/j.gastro.2020.05.048.Search in Google Scholar PubMed PubMed Central
96. Donati Zeppa, S, Agostini, D, Piccoli, G, Stocchi, V, Sestili, P. Gut microbiota status in COVID-19: an unrecognized player?. Front Cell Infect Microbiol 2020;10:576551. https://doi.org/10.3389/fcimb.2020.576551.Search in Google Scholar PubMed PubMed Central
97. Bradley, KC, Finsterbusch, K, Schnepf, D, Crotta, S, Llorian, M, Davidson, S, et al.. Microbiota-driven tonic interferon signals in lung stromal cells protect from influenza virus infection. Cell Rep 2019;28:245–56. https://doi.org/10.1016/j.celrep.2019.05.105.Search in Google Scholar PubMed
98. Miquel, S, Martín, R, Rossi, O, Bermúdez-Humarán, LG, Chatel, JM, Sokol, H, et al.. Faecalibacterium prausnitzii and human intestinal health. Curr Opin Microbiol 2013;16:255–61. https://doi.org/10.1016/j.mib.2013.06.003.Search in Google Scholar PubMed
99. Freeman, EE, McMahon, DE, Lipoff, JB, Rosenbach, M, Kovarik, C, Desai, SR, et al.. The spectrum of COVID-19-associated dermatologic manifestations: an international registry of 716 patients from 31 countries. J Am Acad Dermatol 2020;83:1118–29. https://doi.org/10.1016/j.jaad.2020.06.1016.Search in Google Scholar PubMed PubMed Central
100. Mirza, FN, Malik, AA, Omer, SB, Sethi, A. Dermatologic manifestations of COVID-19: a comprehensive systematic review. Int J Dermatol 2021;60:418–50. https://doi.org/10.1111/ijd.15168.Search in Google Scholar PubMed
101. Genovese, G, Moltrasio, C, Berti, E, Marzano, AV. Skin manifestations associated with COVID-19: current knowledge and future perspectives. Dermatology 2021;237:1–12. https://doi.org/10.1159/000512932.Search in Google Scholar PubMed PubMed Central
102. Jiang, L, Tang, K, Levin, M, Irfan, O, Morris, SK, Wilson, K, et al.. COVID-19 and multisystem inflammatory syndrome in children and adolescents. Lancet Infect Dis 2020;20:e276–88. https://doi.org/10.1016/s1473-3099(20)30651-4.Search in Google Scholar PubMed PubMed Central
103. Rowley, AH. Understanding SARS-CoV-2-related multisystem inflammatory syndrome in children. Nat Rev Immunol 2020;20:453–4. https://doi.org/10.1038/s41577-020-0367-5.Search in Google Scholar PubMed PubMed Central
104. Nakra, NA, Blumberg, DA, Herrera-Guerra, A, Lakshminrusimha, S. Multi-system inflammatory syndrome in children (MIS-C) following SARS-CoV-2 infection: review of clinical presentation, hypothetical pathogenesis, and proposed management. Children 2020;7:69. https://doi.org/10.3390/children7070069.Search in Google Scholar PubMed PubMed Central
105. Mongioì, LM, Barbagallo, F, Condorelli, RA, Cannarella, R, Aversa, A, La Vignera, S, et al.. Possible long-term endocrine-metabolic complications in COVID-19: lesson from the SARS model. Endocrine 2020;68:467–70. https://doi.org/10.1007/s12020-020-02349-7.Search in Google Scholar PubMed PubMed Central
106. Dutta, S, Sengupta, P. SARS-CoV-2 and male infertility: possible multifaceted pathology. Reprod Sci 2021;28:23–6. https://doi.org/10.1007/s43032-020-00261-z.Search in Google Scholar PubMed PubMed Central
107. Brugliera, L, Spina, A, Castellazzi, P, Cimino, P, Arcuri, P, Negro, A, et al.. Nutritional management of COVID-19 patients in a rehabilitation unit. Eur J Clin Nutr 2020;74:860–3. https://doi.org/10.1038/s41430-020-0664-x.Search in Google Scholar PubMed PubMed Central
108. Caccialanza, R, Laviano, A, Lobascio, F, Montagna, E, Bruno, R, Ludovisi, S, et al.. Early nutritional supplementation in non-critically ill patients hospitalized for the 2019 novel coronavirus disease (COVID-19): rationale and feasibility of a shared pragmatic protocol. Nutrition 2020;74:110835. https://doi.org/10.1016/j.nut.2020.110835.Search in Google Scholar PubMed PubMed Central
109. Sosnowski, K, Lin, F, Mitchell, ML, White, H. Early rehabilitation in the intensive care unit: an integrative literature review. Aust Crit Care 2015;28:216–25. https://doi.org/10.1016/j.aucc.2015.05.002.Search in Google Scholar PubMed
110. Simpson, R, Robinson, L. Rehabilitation after critical illness in people with COVID-19 infection. Am J Phys Med Rehabil 2020;99:470–4. https://doi.org/10.1097/phm.0000000000001443.Search in Google Scholar PubMed PubMed Central
111. Masiero, S, Zampieri, D, Del Felice, A. The place of early rehabilitation in intensive care unit for COVID-19. Am J Phys Med Rehabil 2020;99:677–8. https://doi.org/10.1097/phm.0000000000001478.Search in Google Scholar
© 2022 Walter de Gruyter GmbH, Berlin/Boston
Articles in the same Issue
- Frontmatter
- Minireview
- The effect of endurance, resistance training, and supplements on mitochondria and bioenergetics of muscle cells
- Reviews
- Cellular cross talk between epicardial fat and cardiovascular risk
- The physiological insight of Coenzyme-Q10 administration in preventing the incidence of reperfusion arrhythmia among patients undergoing coronary artery bypass grafting surgery
- Is it possible to treat nonalcoholic liver disease using a flavanol-based nutraceutical approach? Basic and clinical data
- An overview of post COVID sequelae
- Clinical implications of anti-idiotype antibodies in COVID-19
- Original Articles
- Monocyte chemoattractant protein-1 levels are associated with major depressive disorder
- Detection of soluble suppression of tumorigenicity 2 and N-terminal B-type natriuretic peptide in a rat model of aortic regurgitation: differential responses to omecamtiv mecarbil
- Preoperative monocyte-to-lymphocyte ratio as a potential predictor of bladder cancer
- AICAR promotes endothelium-independent vasorelaxation by activating AMP-activated protein kinase via increased ZMP and decreased ATP/ADP ratio in aortic smooth muscle
- Moderate-intensity exercise decreases the circulating level of betatrophin and its correlation among markers of obesity in women
- Impact of muscle mass on blood glucose level
- Behavioral alterations, brain oxidative stress, and elevated levels of corticosterone associated with a pressure injury model in male mice
- Patterns of drug therapy, glycemic control, and predictors of escalation – non-escalation of treatment among diabetes outpatients at a tertiary care center
- Pattern, severity, and outcome of adverse drug reactions in a tertiary care hospital: an evidence from a cross-sectional study
Articles in the same Issue
- Frontmatter
- Minireview
- The effect of endurance, resistance training, and supplements on mitochondria and bioenergetics of muscle cells
- Reviews
- Cellular cross talk between epicardial fat and cardiovascular risk
- The physiological insight of Coenzyme-Q10 administration in preventing the incidence of reperfusion arrhythmia among patients undergoing coronary artery bypass grafting surgery
- Is it possible to treat nonalcoholic liver disease using a flavanol-based nutraceutical approach? Basic and clinical data
- An overview of post COVID sequelae
- Clinical implications of anti-idiotype antibodies in COVID-19
- Original Articles
- Monocyte chemoattractant protein-1 levels are associated with major depressive disorder
- Detection of soluble suppression of tumorigenicity 2 and N-terminal B-type natriuretic peptide in a rat model of aortic regurgitation: differential responses to omecamtiv mecarbil
- Preoperative monocyte-to-lymphocyte ratio as a potential predictor of bladder cancer
- AICAR promotes endothelium-independent vasorelaxation by activating AMP-activated protein kinase via increased ZMP and decreased ATP/ADP ratio in aortic smooth muscle
- Moderate-intensity exercise decreases the circulating level of betatrophin and its correlation among markers of obesity in women
- Impact of muscle mass on blood glucose level
- Behavioral alterations, brain oxidative stress, and elevated levels of corticosterone associated with a pressure injury model in male mice
- Patterns of drug therapy, glycemic control, and predictors of escalation – non-escalation of treatment among diabetes outpatients at a tertiary care center
- Pattern, severity, and outcome of adverse drug reactions in a tertiary care hospital: an evidence from a cross-sectional study