Cardiovascular disease and infections are predominant causes of death in patients with kidney disease. Whereas cardiovascular disease is a primary focus of many research activities in nephrology, awareness, and funding, research on aberrant immune function and infections in patients with kidney disease is sparse. Recently, the coronavirus disease 2019 (COVID-19) pandemic garnered more attention for kidney disease as a risk factor for severe and lethal COVID-19 and that kidney disease can impair the immune response to vaccines.1 However, the molecular mechanisms underlying the increased susceptibility to severe infections and impaired vaccine responses remain unclear, as do possible therapeutic interventions to interfere with the effect of kidney disease on the immune system.
Kidney disease is associated with a state of secondary immunodeficiency (SID), here referred to as SID related to kidney disease (SIDKD). Notably, most chronic organ failures, but also malnutrition, aging, chronic infections, and numerous immunosuppressive drugs, can cause SID (Figure 1). In this review, we highlight the unmet needs relating to SIDKD in terms of awareness, a new definition, epidemiology, pathophysiologic mechanisms, and a call for targeting SIDKD with specific interventions to improve outcomes in patients with kidney disease.
Figure 1.: Factors leading to secondary immmunodeficiency. Multiple factors contribute to SIDKD, including HIV, most forms of chronic organ failure (such as heart, liver, and kidney failure), malnutrition, aging, cancer, dysbiosis, gut disorders, chronic infections (such as tuberculosis), and various immunosuppressive drugs.
Toward a New and Uniform Definition of SIDKD
A definition of SIDKD has not been established but would be essential for epidemiologic studies, preclinical science, clinical research, and teaching. Ideally, such a definition would match those used for other forms of immunodeficiency (ID). The European Society for Immunodeficiencies working definitions for the heterogeneous inborn errors of immunity, and definitions of SID used in other medical contexts, provide some guidance.2,3 In general, such definitions include a clinical component indicating increased susceptibility to infections, represented by recurrent or severe infections and/or insufficient vaccine responses. A second component refers to abnormal results of immunophenotyping. For SIDKD, any form of kidney disease, but particularly CKD, would be a conditio sine qua non. Hence, we propose the a definition of SIDKD in Table 1, which is shown in a simplified way in Figure 2A. Of note, kidney disease can also evolve from primary ID. For example, genetic complement deficiencies and defects in phagocytosis or lymphocyte apoptosis may first lead to systemic autoimmunity, followed by immune complex GN. Furthermore, in patients with primary ID, systemic infections, kidney infections, or recurrent use of nephrotoxic antimicrobial drugs may trigger kidney disease (Figure 2B).
Figure 2.: Definition and the path toward SIDKD. (A) Kidney disease, i.e., CKD, is associated with SIDKD by ultimately increasing the susceptibility to infections, as represented by recurrent or severe infections and/or insufficient vaccine responses. Such abnormalities also contribute to defective humoral and cellular immune responses. (B) Primary ID can also cause kidney disease, e.g., genetic complement deficiencies and defects in phagocytosis or lymphocyte apoptosis that may lead to systemic autoimmunity and immune complex GN. In patients with primary ID, systemic or kidney infections and recurrent use of nephrotoxic antimicrobial drugs may trigger kidney disease. Thus, all risk factors contributing to SIDKD are associated with increased susceptibility to infection, impaired vaccine response, and attenuated sterile inflammation.
Table 1. -
Definition of SIDKD
Domain |
Criterion |
Clinical |
|
Kidney disease |
CKD (as defined by KDIGO) |
AND any of the following |
Infection |
Severe infection requiring antimicrobial treatment and hospitalization |
|
OR Repetitive infections requiring repetitive antimicrobial treatment or permanent antimicrobial prophylaxis |
|
OR Persistent infection requiring permanent or repetitive antimicrobial treatments |
|
OR Opportunistic infections |
OR Impaired response to vaccines |
Lack of developing an antigen-specific immune response to vaccines that produce a high rate of immune response in healthy individuals |
OR Attenuated sterile inflammation |
For example, declining activity of autoimmune diseases or lower incidence of acute gouty arthritis than expected given the profound and persistent hyperuricemia |
Laboratory |
|
AND/OR Humoral |
Subnormal serum levels of total Ig or Ig subclasses or IgG subclasses |
|
OR Deficiency in complement factors |
AND/OR Cellular |
OR Deficiency of any other humoral effector element of immunity |
|
Subnormal white blood count, neutropenia, lymphopenia, or any specific lymphocyte subset, such as B cells or CD4 T cells |
|
OR Abnormal results in immune cell activation assays and phagocytosis |
KDIGO, Kidney Disease Improving Global Outcomes.
Epidemiology of SIDKD
Without a uniform definition of SIDKD, definite epidemiologic data have remained vague. One approach is to assess infection rates in CKD/ESKD patient cohorts, e.g., by using hospitalization and mortality data.
Impaired Host Defense to Infections
The global influence of COVID-19 has raised awareness of the vulnerability of patients with CKD/ESKD to lethal viral infection.45–6 Cohort studies observed a COVID-19 mortality rate of 17%–44% among the CKD/ESKD population, particularly in those requiring dialysis across countries such as Italy,7,8 Spain,9,10 Korea,11 Turkey,12 Germany,13 the United States,14,15 Sweden,16 and Australia/New Zealand.17 Kidney transplant recipients had an even higher mortality of COVID-19 (37.5%; hazard ratio, 3.36; 95% confidence interval [95% CI], 1.19 to 9.50, P=0.022) compared with patients with ESKD on dialysis (11.3%).18
Infection-associated mortality rates have also been reported in large CKD outcome trials (Table 2). For example, the Dapagliflozin in Patients with Chronic Kidney Disease trial reported an infection-related mortality rate of 18.6% in >2000 patients with CKD who did and did not have diabetes.19 In the Atherosclerosis Risk in Communities Study, involving >9000 patients in the United States from 1996 to 2011, CKD stage G5 was associated with an adjusted hazard ratio of 3.76 (95% CI, 1.48 to 9.58) for infection-related death.20 Retrospective cohort studies before the pandemic reported a higher infection-related mortality in patients on dialysis compared with kidney transplantation recipients from the United States (adjusted death rate of 18.6 versus 12.8 per year; age 60–64)21 and the European Renal Association–European Dialysis and Transplant Association registry (82-fold in patients on dialysis versus 32-fold in transplant recipients).22 Data from the United States in 2016 report an incidence of 614 hospitalizations per 1000 person-years in patients with CKD,23 which places infections as the second leading cause for hospitalization. A lower eGFR is associated with an increased one- to three-fold risk of hospitalization as CKD stages progress.20,24,25 In >230,000 Canadian patients, the risk for hospitalization due to community-acquired pneumonia increased with decreasing eGFR.26 The influenza A virus subtype H1N1 (swine influenza),27 severe acute respiratory syndrome,28,29 and tuberculosis30,31 are associated with a longer duration in the hospital and a more aggressive clinical course or death in patients receiving dialysis, compared with the general population (Table 2). Similar observations have been documented for parasitic infections, e.g., Plasmodium falciparum or Plasmodium vivax32 and protozoa33 in patients with CKD in developing countries.
Table 2. -
Selected epidemiologic studies of SIDKD
Consequences of SIDKD |
Clinical study |
Mortality, Incidence, or Prevalence |
Reference |
Infection |
General |
|
|
|
DAPA-CKD study |
22% of non-CV death without TDM2 and 17.8% with TDM2 (HR, 0.64; 95% CI, 0.36 to 1.16) |
19
|
Stockholm CREAtinine Measurement Project |
Community-acquired infection in patients with eGFR <30ml/min per 1.73 m2 (IRR, 1.53; 95% CI, 1.39 to 1.69) and eGFR 30-59/min per 1.73 m2 (IRR, 1.08; 95% CI, 1.01 to 1.14) |
202
|
ASN 2003 USRDS |
CKD infection rate per 100 patients per year: sepsis, 4; UTI, 22; pneumonia, 14; any infection, 33; two-fold higher in patients with ESKD than in those with CKD |
203
|
CanPREDDICT prospective cohort study (Canada) |
Risk of infection in 24.3% patients with eGFR 15–45 ml/min per 1.73 m2 (HR of mortality, 3.39; 95% CI, 2.65 to 4.33) |
204
|
The Atherosclerosis Risk in Communities Study |
HRs for infection-related death: eGFR 60–89 ml/min per 1.73 m2, 0.99 (95% CI, 0.80 to 1.21); eGFR 30–59 ml/min per 1.73 m2, 1.62 (95% CI, 1.20 to 2.19); eGFR 15–29 ml/min per 1.73 m2, 3.76 (95% CI, 1.48 to 9.58) |
20
|
Propensity score–matched retrospective cohort study (Canada) |
Increased risk for infection-related hospitalization PD versus HD with HR of 1.52 (95% CI, 1.38 to 1.68) |
205
|
Observational cohort study (Australia, New Zealand) |
Incidence rates of infectious mortality for PD 2.8 and for HD 1.7 per 100 patients per year (IRR PD versus HD, 1.66, 95% CI, 1.47 to 1.86) |
206
|
COVID-19 |
Mexican Open Registry of patients with COVID-19 |
Mortality 20.8% higher in patients with diabetes and CKD versus those with CKD only |
207
|
Retrospective study (UK Transplant Center) |
Overall infection-related mortality, 0.4% |
208
|
ERACODA retrospective cohort study |
Mortality in kidney transplant recipients (16.9%) and patients on HD (23.9%) within 28 days; HR, 1.78 (95% CI, 1.22 to 2.61) |
209
|
Systemic review and meta-analysis (Taiwan) |
Overall mortality rate of 22.4% in patients on HD with COVID-19 (95% CI, 17.9% to 27.1%) |
210
|
Retrospective cohort study (United States) |
Overall mortality of 24.9% in patients on HD with COVID-19 |
211
|
Multicenter retrospective observational study (Turkey) |
Overall mortality of 16.3% in patients on HD with COVID-19 |
212
|
Retrospective cohort study (Spain) |
Mortality of 36% in patients with CKD stage G5D |
10
|
Tuberculosis |
Tuberculosis register (United Kingdom) |
TB incidence among patients with CKD per year: CKD stage G1–3, 80 per 100,000; CKD stage G4–5, 128 per 100,000; CKD stage G5D, 256 per 100.000 (95% CI, 183 to 374) |
213
|
Population-based study (British Columbia) |
Relative risk of TB in patients on HD is 25.3% (95% CI, 22.86 to 31.49) |
214
|
Systemic review and meta-analysis |
OR of 3.62 for TB in patients on HD (95% CI, 1.79 to 7.33) |
215
|
Retrospective cohort study (Japan) |
Adjusted OR for TB-related death: eGFR <30 ml/min per 1.73 m2, 2.99 (95% CI, 1.20 to 7.51) |
216
|
Retrospective cohort study (Taiwan) |
Adjusted OR of TB 1.45-fold higher in patients with CKD versus those without CKD (95% CI, 1.27 to 1.64) |
217
|
Herpes zoster |
Taiwan Longitudinal Healthy Insurance Database |
Risk for herpes zoster in patients with CKD: HR of 1.6 (95% CI, 1.41 to 1.81) |
218
|
Clostridium difficile infection |
Single-center, retrospective, case-control study (Korea) |
Increased risk for CDI and higher mortality in CKD stage G4–5 (OR, 2.9) and ESKD G5D (OR, 3.34) |
219
|
Case-control study (United States) |
OR of CDI-associated diarrhea was 2.60 (95% CI, 1.25 to 5.41) in ESKD versus 1.07 (95% CI, 0.65 to 1.77) in CKD stage G3–5 |
220
|
Case-control study (Mexico) |
3-month mortality rate 10.4%; CDI in 29.6% of patients with CKD |
221
|
Case-control study (Poland) |
CDI in 67% of patients with CKD stage G5D versus 5.7% in those with CKD stage G1 |
222
|
Case-control study (Korea) |
OR of 4.44 (95% CI, 2.19 to 8.99) in CKD stage G5D |
223
|
Pneumonia |
Community-based study Alberta Kidney Disease Network (Canada) |
Adjusted HR for hospitalization with pneumonia: eGFR 45–59 ml/min per 1.73m2, 3.23 (95% CI, 2.40 to 4.36); eGFR 33–44 ml/min per 1.73 m2, 9.67 (95% CI, 6.36 to 14.69); eGFR <30 ml/min per 1.73 m2, 15.04 (95% CI, 9.64 to 23.47) |
26
|
Cardiovascular Health Study (United States) |
Infection-related hospitalization: eGFR 60–89 ml/min per 1.73 m2, 16% eGFR 45–59 ml/min per 1.73m2, 37% eGFR 15–44 ml/min per 1.73 m2, 64% |
25
|
Retrospective cohort study (United Kingdom) |
IRR for pneumonia: eGFR 45–59 ml/min per 1.73 m2, 0.95 (95% CI, 0.89 to 1.01); eGFR 30–44 ml/min per 1.73 m2, 1.19 (95% CI, 1.11 to 1.28); eGFR 15–29 ml/min per 1.73m2, 1.73 (95% CI, 1.57 to 1.92); eGFR <15 ml/min per 1.73 m2, 3.04 (95% CI, 2.42 to 3.83). IRR for LRTI: eGFR <15 ml/min per 1.73 m2, 1.47 (95% CI, 1.34 to 1.62) |
224
|
Nationwide population-based study (Taiwan) |
Adjusted HR of pneumonia: HR of 1.97 (95% CI, 1.89 to 2.05) in patients with CKD, HR of 1.4 (95% CI, 1.31 to 1.49) in outpatient pneumonia, 2.17 (95% CI, 2.07 to 2.29) in inpatient pneumonia |
225
|
Observational study (Spain) |
15.8% mortality in patients with CKD versus 8.3% in those without CKD (OR of 0.38 in CKD and pneumonia, 95% CI, 0.04 to 3.05) |
226
|
Retrospective cohort study (China) |
Risk of empyema in 66.7% of patients with CKD and 52.5% of those with ESKD |
227
|
Sepsis |
Retrospective cohort study (France) |
70% mortality rate in patients with CKD after 28 d versus 50% in those without CKD |
228
|
Retrospective cohort study (United Kingdom) |
IRR for sepsis: eGFR <60 ml/min per 1.73 m2, 5.56 (95% CI, 3.90 to 7.94) |
224
|
Observational cohort study (Germany) |
HR of 2.25 (95% CI, 1.46 to 3.46) in patients with CKD versus those without CKD |
229
|
Dialysis catheter– and dialysis process–related infections |
EPIBACDIAL multicenter prospective study (France) |
Incidence of bacteremia in patients on HD of 0.93 episode per 100 patient-months; vascular access RR of 7.6 (95% CI, 3.7 to 15.6) |
38
|
Matched cohort study (United States) |
Decreased all-cause mortality in patients on home HD versus those on PD; HR of 0.80 (95% CI, 0.73 to 0.87) |
230
|
Retrospective cohort study (Algeria) |
Central venous catheter–related infection in patients on HD: 16.6 infections/1000d |
231
|
Systemic review of cohort studies |
Higher RR using catheters compared with fistulas in mortality (RR, 1.53, 95% CI, 1.41 to 1.67) and infections (RR, 2.12, 95% CI, 1.79 to 2.52) |
232
|
Retrospective cohort study (Germany) |
Infection risk of permanent HD catheters: 17.9% (systemic infection, 2.26 episodes per 1000 catheter days; local infection, 0.6 episodes per 1000 catheter days) |
233
|
Retrospective cohort study of outpatient dialysis center (United States) |
Peritonitis due to peritoneal dialysis solution in 14 of 22 patients on PD |
234
|
Retrospective observational cohort study using NHSN surveillance data (United States) |
Bloodstream infection due to buttonhole cannulation to access arteriovenous fistulas in 37% of patients on dialysis; adjusted RR, 2.6, 95% CI, 2.4 to 2.8 |
235
|
The Network 9 peritonitis and catheter survival studies (United States) |
Infection-related cause of catheter removal in 68% of patients; peritonitis and exit/tunnel infections in 13.4% of patients |
236
|
HEMO study; prospective multicenter study (United States) |
35% infection-related hospitalization due to vascular access in 21% of cases; infection-related death, 23% (RR of infection-related death with age 1.47, 95% CI, 1.29 to 1.68) and with comorbidity 1.46, 95% CI, 1.21 to 1.76) |
40
,
237
|
Observational cohort single-center study (United States) |
Patients on PD and HD with similar overall infection rates (0.77 for HD versus 0.86 for PD per year); PD versus HD: RR, 1.3, 95% CI, 0.93 to 1.8); HD catheters increased rate of bacteremia by 67% |
41
|
Case-control study (United Kingdom) |
Incidence of endocarditis in patients with HD and PD due to AVF, 41.3%; DLTC, 37.9%; PTFE, 10.3%; DLNTC, 4% |
238
|
Staphylococcus aureus
|
Meta-analysis of methicillin-resistant Staphylococcus aureus colonization |
Dialysis modality of infection in 7.2% of patients on HD (95% CI, 4.9% to 9.9%) and 1.3% in patients on PD (95% CI, 0.5% to 2.4%) |
239
|
Nationwide study on the Danish National Registry on Regular Dialysis and Transplantation (Denmark) |
Infection in 12.8% of patients on dialysis (RR, 7.42, 95% CI, 5.63 to 9.79) |
240
|
Oral yeast colonization |
Retrospective cohort study (Turkey) |
Incidence of oral yeast colonization was 32.1% in RTR, 40% in HD, 20.9% in CAPD, and 18% in healthy controls |
241
|
Drug-related infection |
Retrospective cohort study in patients with AAV (China) |
Pulmonary infection after rituximab treatment: 20.9 per 100 person-years (HR, 1.493, 95% CI, 1.017 to 2.191) |
242
|
Attenuated sterile inflammation |
Gout |
NHANES studies of gout in patients with CKD and marked hyperuricemia (United States) |
Prevalence of gout: CKD stage G1, 4%; CKD stage G2, 6%–10%; CKD stage G3, 11%–13%; CKD stage >G4, 30% (95% CI, 1.94 to 5.24 to gout patients without CKD) |
64
|
GCKD study of gout in patients with CKD and marked hyperuricemia (Germany) |
Prevalence of gout: eGFR ≥60 ml/min per 1.73 m2, 16%; eGFR <30 ml/min per 1.73 m2, 35.6% (prevalence ratio 1.46, 95% CI, 1.21 to 1.77) |
65
|
Impaired vaccine response |
COVID-19 vaccine |
Cohort study (Israel) |
Lower antibody levels in patients on HD versus controls (OR of 2.7, 95% CI, 1.13 to 7.51) |
50
|
Cohort study (Germany) |
4.92% nonhumoral responders and 28.4% noncellular responders in patients on HD who were fully vaccinated with BTN162b2 |
45
|
Cohort study (United States) |
Impaired humoral immune response to the COVID-19 Ad26.COV2.S vaccine in 33.3% of patients on HD |
46
(preprint)
|
Cohort study (Germany) |
Impaired humoral immune response to the COVID-19 BTN162b2 vaccine in patients on HD and kidney transplant recipients |
47
|
Cohort studies (Germany) |
Impaired humoral and cellular immune response after BTN162b2 vaccination in kidney transplant recipients |
48
,
49
|
Hepatitis B vaccine |
Cohort study (Brazil) |
Vaccine response to HBV was 70% in patients on HD (anti-HBs seroconversion, OR, 5.239, 95% CI, 1.279 to 21.459) |
54
|
Case-control study (India) |
Seroconversion rates after 3 doses of HBV vaccine: 87.5% in mild CKD, 66.6% in moderate CKD, 35.7% in HD |
55
|
Prospective cohort study (Spain) |
1 month after vaccination, 77.5% of patients had seroconverted, 72.5% achieved high antibody response, whereas 22.5% were nonresponders |
56
|
Placebo-controlled, randomized, double-blind trial (France) |
Immune response observed in 60% of patients on HD after vaccination |
57
|
Diphtheria and tetanus vaccines |
Prospective cohort study (Germany) |
Anti-diphtheria antibody level after 5 years in 32% of patients on HD; anti-tetanus antibody level after 5 years in 65% of patients on HD |
59
|
Prospective cohort study (Germany) |
Overall protection rate against diphtheria: 22% |
60
|
Pneumococcal vaccine |
Prospective cohort study (Germany) |
Postvaccine antibody titers in 83% of patients with CKD after 1 month, 68% after 6 months, 48% after 1 year |
61
|
Influenza vaccine |
Prospective cohort study |
66% protection rate against A-H3N2, 25% against A-H1N1, and 27% against B strain in patients on HD; booster injection had low effect |
62
|
DAPA-CKD, Dapagliflozin in Patients with Chronic Kidney Disease; CV, cardiovascular; TDM2, type 2 diabetes mellitus; IRR, incidence rate ratio; ASN, American Society of Nephrology; USRDS, United States Renal Data System; UTI, urinary tract infection; CanPREDDICT, Canadian Study of Prediction of Death, Dialysis and Interim Cardiovascular Events; HR, hazard ratio; ERACODA, European Renal Association COVID-19 Database; TB, tuberculosis; OR, odds ratio; CDI, Clostridium difficile infection; LRTI, lower respiratory tract infection; RR, risk ratio; NHSN, National Healthcare Safety Network; HEMO, Hemodialysis Study; AVF, arteriovenous fistula; DLTC, dual-lumen tunneled catheter; PTFE, polytetrafluoroethylene; DLNTC, dual-lumen nontunneled catheter; RTR, renal transplant recipients; CAPD, continuous ambulatory peritoneal dialysis; AAV, ANCA-associated necrotizing vasculitis; NHANES, National Health and Nutrition Examination Survey; GCKD, German Chronic Kidney Disease; HBV, hepatitis B virus.
Dialysis-related infections can occur upon recurrent insertion of arteriovenous fistulas/grafts or dialysis catheters.34,35 The type of dialysis access matters in this context.36,37 Patients undergoing dialysis via catheters have a two-fold higher risk for bacteremia or sepsis38,39 and infection-related death40 compared with those with arteriovenous fistulas/grafts. In addition, patients on hemodialysis (HD) are more susceptible to bacteremia, whereas patients on peritoneal dialysis (PD) have an increased risk for peritonitis.36,41,42 Furthermore, contamination of dialysis equipment and dialysate43,44 with diverse virulent microorganisms42,43 can underlie SIDKD in this population.
Impaired Immune Responses to Vaccines
Another clinical indicator of SIDKD is impaired vaccine response. Data from the ongoing COVID-19 pandemic show that patients on HD and PD may have a humoral antibody response against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) after receipt of the mRNA vaccine BNT162b2.45,46(preprint)–49 However, the levels of anti-S1 IgG antibodies and SARS-CoV-2 surrogate neutralizing antibodies were substantially lower in patients on dialysis after the first and second vaccination compared with patients without CKD.50,51 Within 12 weeks surrogate neutralizing antibodies start to decrease from 93% to 85% in patients on PD and from 87% to 79% in patients on HD, respectively.52–53–53 Consistently, diminished seroconversion rates after immunization have been reported for hepatitis B virus (no difference in responsiveness between patients on PD and HD),54555657–58 diphtheria and tetanus,59,60 pneumococcal,61 and influenza62 vaccines compared with the general population. As vaccine-induced antibody levels and humoral immunity decrease, patients with ESKD benefit from booster vaccinations.63
Sterile Forms of Inflammation
SIDKD also implies an attenuation of noninfectious forms of inflammation, such as the autoimmune disease activity that declines as CKD progresses. In addition, patients with CKD/ESKD who have significant hyperuricemia show an unexpectedly low rate of acute gout attacks.64,65
Although the lack of a common definition of SIDKD makes definite epidemiologic data scarce, numerous clinical observations suggest that SIDKD is prevalent in patients with CKD and ESKD; therefore, mechanistic explanations of SIDKD are required.
Immunophenotype of Patients with Kidney Disease
Biomarkers for SID in clinical use include white blood cell count, total Ig levels, measurement of antibodies to specific pathogens or vaccine responses, and fluid-phase complement factors, but also specific assays of neutrophil and T cell function. Immunophenotyping of patients with CKD/ESKD reveals signs of chronic inflammation and impaired immune effector functions (Figure 3A).66
Figure 3.: Immunophenotype in kidney disease. (A) SIDKD is associated with an abnormal immunophenotype, characterized by chronic inflammation as a result of persistent immune cell activation, and simultaneously with immune paralysis due to impaired immune effector functions. (B) Mechanisms that are involved in SIDKD include suppressed innate immune responses, e.g., impaired leukocyte and platelet function, reduced antigen-presenting ability of macrophages and dendritic cells to T and B cells, and impaired adaptive immune responses (e.g., T lymphocyte maturation and activation, reduced antibody production by plasma cells).
Impaired Function of Innate Immunity
In patients with CKD/ESKD, biomarkers of oxidative stress, such as advanced oxidation protein products and myeloperoxidase, and for inflammation, such as high sensitivity C-reactive protein and IL-6, are inter-related.67,68 Thus, uremia leads to chronic low-grade inflammation, in which permanent immune cell activation and secretion of inflammatory cytokines, together with uremic solutes/metabolites, reduce the ability of neutrophils and monocytes to migrate to the inflammation site (Figure 3B).6970–71 Mechanistically, selectin-induced slow leukocyte rolling and transmigration are abolished,72,73 and β2 integrins are downregulated.74 Moreover, neutrophils from patients with ESKD are more likely to undergo apoptosis, which decreases the ability of neutrophils to form neutrophil extracellular traps 75,76 and to phagocytose and kill pathogens,77 mechanisms that are associated with an impaired host defense in SIDKD. A number of uremic solutes/metabolites that affect neutrophil oxidative burst have been identified, such as phenylacetic acid, resistin, and methylglyoxal.78,79
Monocytes from patients on PD are hyporeactive to LPS stimulation and release fewer proinflammatory cytokines in comparison to control subjects.80,81 Monocytes and monocyte-derived dendritic cells display decreased endocytosis and impaired maturation when cultured in uremic serum or when obtained from patients with ESKD.82 Moreover, dendritic cells and macrophages have a reduced antigen-presenting capability to activate T cells, which might be due to an abnormal expression of Toll-like receptor 483 and costimulatory molecules CD80 and CD86.84 An increased intracellular uptake of uric acid, related to an impaired renal clearance of uric acid uptake, contributes to uremia-related monocyte dysfunction.74
In patients on HD, higher serum levels of mannose-binding lectin are associated with an increased risk of severe infection.85,86 Mannose-binding lectin can bind to carbohydrates on the surfaces of bacteria and viruses (e.g., SARS-CoV-287) and initiate complement-driven pathogen lysis.88,89 However, uncontrolled complement activation also contributes to hyperinflammation, cell injury, immunothrombosis, and multiorgan failure during COVID-19 infection.90 In addition, monocytes/macrophages from patients with ESKD have increased expression and activity of the macrophage scavenger receptors SR-A and CD36,9192–93 but decreased inducible nitric-oxide synthase expression94 and phagocytic capability, e.g., of peritoneal macrophages in chronic PD and PD-induced peritonitis95,96 and potentially also in alveolar macrophages in the context of pulmonary infections. ESKD reduces the cell number and cytotoxicity of natural killer (NK) cells in association with downregulation and modulation of ligand expression for activating receptors on NK cells.97,98 Thus, uremia impairs the functional properties of neutrophils, monocyte/macrophages, and NK cells that are important to maintain a sufficient host defense.
Impaired Platelet Function
Beyond their important role in homeostasis, platelets are important cellular contributors to host defense99 and essential for sufficient neutrophil activation and recruitment into peripheral organs under different inflammatory conditions. However, gut microbial uremic toxins, which accumulate during CKD,100 and altered HDL levels contribute to altered homeostasis and immune effector functions in SIDKD (Figure 3).101 This dichotomy may be explained by the fact that critical factors determining the effective formation of platelet-leukocyte aggregates, e.g., plasma fibrinogen levels involved in bond formation between integrins, GPIIbIIIa on platelets, and Mac-1 on neutrophils, are increased in patients with CKD.102 Furthermore, platelets release more α-granules on stimulation, leading to increased incorporation and expression of P-selectin on the platelet surface, which, in turn, binds to PGSL-1, which is expressed on various leukocyte subsets, including neutrophils.103 Thus, uremic platelet dysfunction not only promotes bleeding complications104 but also immunothrombosis in CKD.105,106
Impaired Function of Adaptive Immunity
Impaired vaccine responses indicate that CKD/ESKD also suppresses adaptive immunity (Figure 3B).107 This suppression involves aberrant T-cell activation caused by altered dendritic cell and macrophage antigen presentation and increased apoptosis of effector CD4+ and CD8+ T cells.98,108,109 Studies performed in vitro show decreased T helper (Th) CD4 cell proliferation in the uremic milieu.110,111 HD and PD have different effects on Th cell phenotypes and proliferation.112,113 In patients on PD, the maturation of both effector Th1 and Th2 CD4 cell subsets is impaired, and there are increased numbers of central memory T cells and CD8+ naive T cells compared with controls and patients on HD.113 In contrast, patients on HD seem to have sustained Th cell maturation, but the immune response is mainly driven via Th1 CD4 (e.g., production of IFN-γ) rather than Th2 CD4 cells.114,115 A possible explanation for the increased Th1/Th2 ratio in patients on HD could be increased IL-12 production.115 Moreover, the different clinical course of infection, e.g., hepatitis B virus, between patients on dialysis and the general population relates to dysfunctional CD8 cytotoxic T cells, which are needed to eliminate infected cells, and dysfunctional CD4 T cells, which sustain CD8 cytotoxic T cells and induce antibody production from B and plasma cells.103
Patients with CKD/ESKD show a defective or repressed humoral and vaccination response.116 B-cell lymphopenia, due to increased B-cell apoptosis and insufficient B-cell proliferation, is common in patients on HD and controls.117118–119 This has been related to decreased Bcl-2 expression and a resistance to IL-7 and B cell–activating factor BAFF/BLyS.117,120 Patients on HD have fewer B1 cells secreting IgM and IgA and B2 cells producing IgG.106,120 Patients on dialysis and kidney transplant patients have poor antibody, memory B cell, and plasmablast responses to vaccines, such as the COVID-19 (BNT162b2),47 hepatitis B virus, and influenza vaccines compared with the general population (Table 2)121,122. Therefore, these patients may not be sufficiently protected against the respective pathogens. CKD and ESKD also impair adaptive immune responses, which are important for host defense but are also implicated in auto- and alloimmunity.
Pathogenic Mechanisms of SIDKD
SIDKD due to Intestinal Barrier Dysfunction
Evidence indicates that the elevated circulating endotoxin/LPS levels and markers of systemic inflammation in patients with CKD stage G3–5 and 5D are caused by a leaky intestinal barrier and enhanced endotoxin translocation from the intestinal lumen into the circulation.123,124 LPS levels were the highest in patients on HD/PD; levels were similar to those in patients with severe liver disease, irradiation-induced intestinal damage, and decompensated heart failure.123 Elevated LPS levels are considered a strong and independent predictor of mortality.123 Wang et al.125 demonstrated that experimental uremia in rats increases bacterial translocation from the gut into mesenteric lymph nodes, liver, and spleen, which was associated with higher levels of serum IL-6 and C-reactive protein.126 The aberrant humoral and cellular immunity in CKD is partially reversible by treatment with antibiotics that eliminate the intestinal flora.127 Endotoxin tolerance and the see-saw phenomenon in sepsis posit that persistent exposure to bacterial endotoxins induces negative regulators of innate immunity that paralyze the innate and adaptive immune system.127,128 In accordance with this concept, CKD-related intestinal dysbiosis, barrier dysfunction, and bacterial translocation account for the state of persistent systemic inflammation in CKD (Figure 4).129
Figure 4.: Pathomechanisms of SIDKD. Kidney disease is associated with an impaired urinary clearance of immunoregulatory metabolites, increased production of immunoregulatory proteins and persistent immune activation, extrinsic immunosuppressors, and a leaky gut and shift in the secretome of the intestinal microbiota—all of which contribute to SIDKD. The pathogenic mechanisms of SIDKD include dysregulation of innate and adaptive immune responses, such as a decreased phagocytic capability of immune cells to clear pathogens, reduced respiratory burst (ROS production) to kill bacteria and viruses, enhanced immune cell activation (upregulation of cell surface receptors) and release of proinflammatory cytokines (e.g., IL-1β, IL-6, TNFα) by innate immune cells (e.g., neutrophils, monocytes, macrophages, dendritic cells), increased neutrophil apoptosis but decreased neutrophil extracellular trap (NET) formation, and reduced antigen-presenting ability of macrophages and dendritic cells to activate T and B cells, which results in decreased T-cell proliferation and antibody production by plasma cells. Although leukocyte/lymphocyte interactions with endothelial cells are increased, the ability of leukocytes/lymphocytes to migrate is impaired due to a downregulation of, e.g., β 2 integrins. Subsequently, the dysregulation of the innate and adaptive immune system contributes to bacterial overgrowth, infections, and attenuated sterile inflammation.
SIDKD due to Persistent Inflammation
Persistent inflammation is an important trigger of immune paralysis and SID. Indeed, dendritic cells, neutrophils, monocytes, macrophages, and mast cells remain unresponsive to a secondary endotoxin challenge due to an induced suppression of signaling pathways contributing to inflammation. This see-saw phenomenon of immune activation is an important, evolutionarily conserved mechanism that avoids a potentially lethal cytokine storm and septic shock. In patients with permanent triggers of inflammation, this see-saw phenomenon begins at the single-cell level.130 The flood of new immune cells produced daily results in a state of concomitant systemic inflammation and immune paralysis at the organismal level.
The endotoxin tolerance–like concept explains the immunosuppressive state in sepsis, cystic fibrosis, pancreatitis, and tuberculosis.131 In the early phase, cytokine storm–like hyperimmunoreactivity predominates and may be lethal. At later stages, and in chronic disorders, hypoimmunoreactivity (as a sign of SID) prevails, leading to potentially fatal secondary infections.132 Cellular mechanisms that underlie immunosuppression include unresponsiveness of monocytes to LPS challenge ex vivo;133 reactivation of adaptive immune cells, such as T regulatory cells and myeloid-derived suppressor cells134,135; but loss of CD4+ and CD8+ T cells (Figure 4).136,137 However, this induced immune paralysis is not restricted to sepsis or cystic fibrosis, but is also present in other forms of sterile inflammation, e.g., in hepatic and kidney ischemia, coronary occlusion, acute coronary syndromes, and even cancer.138139140–141
An impaired intestinal barrier is a possible source of persistent exposure to bacterial endotoxins, as indicated by increasing endotoxin serum levels at later stages of CKD.123,127
Shifts in the Secretome of the Intestinal Microbiota
The secretome of the intestinal microbiota is an integral component of human physiology and changes in the microbiota composition, which affect the secretome, disturb physiology and homeostasis.142143–144 Kidney disease–related alkalosis, intestinal wall congestion, azotemia, and other metabolic disturbances alter the composition of the intestinal microbiota and hence its secretome.127,145 Among many other aspects of physiology, a uremia-related altered intestinal secretome implies changes in numerous immunoregulatory metabolites, including a reduction in short-chain fatty acids, such as butyrate and acetate, that usually suppress inflammation (Figure 4).146,147 Other gut-derived metabolites that affect the immune system include indoxyl sulfate, p-cresyl sulfate, indole-3-acetic acid, and phenylacetylglutamine.147,148 Mechanistically, indoxyl sulfate and p-cresyl sulfate are associated with systemic inflammation and an altered host defense in patients with CKD, e.g., by inducing glutathione peroxidase, TNFα, and IL-6 release in monocytes149; impairing respiratory burst activity and phagocytosis in neutrophils; and causing eryptosis (programmed death of red blood cells) partly by increasing cytosolic calcium.150
A high salt diet can reduce the intestinal population of Lactobacillus murinus, which decreases indole-3-lactate levels, affecting Th17 cell function in a way that can aggravate experimental hypertension and multiple sclerosis.151 It is possible that CKD or CKD-related medications, dietary restrictions, and use of antibiotics alter the intestinal microbiota,152 leading to concomitant systemic inflammation and ID. Moreover, fecal transplantation in CKD can induce metabolic changes that lead to sarcopenia and immune dysregulation. Mice receiving a fecal transplant from CKD patients had significantly higher plasma trimethylamine N-oxide levels and a different gut microbiota composition than mice receiving a fecal transplant from non-CKD patients.153 These findings suggests that CKD modifies the composition of the intestinal microbiota and its secretome, which, together with barrier dysfunction and bacterial translocation, might explain the persistent systemic inflammation and defective host defense associated with CKD.
Impaired Urinary Clearance of Immunoregulatory Metabolites
Low-molecular-weight solutes involved in both systemic and organ-specific metabolism may impair urinary clearance and exhibit increased production, or decreased breakdown, in kidney disease (Figure 4). For example, p-cresyl sulfate induces an increase in oxidative burst and phagocytosis in human macrophages, but decreases their antigen-presenting ability to T cells by downregulating HLA-DR and CD86.154 Moreover, indoxyl sulfate promotes leukocyte-endothelial interactions through the upregulation of vascular endothelial adhesion molecules, such as E-selectin, via JNK- and oxidative stress–dependent pathways155; can induce the expression of adhesion molecules, such as MAC-1; and increase production of reactive oxygen species (ROS) in a manner dependent on NADPH oxidase and p38 MAPK in monocytes.156 In patients with common variable ID that present with defective B cell to plasma cell development, several studies report a strong association between trimethylamine N-oxide and asymmetric dimethylarginine with systemic inflammation, immune cell activation, and gut microbial abundance.157158–159
Whereas the above-mentioned metabolites drive systemic inflammation, CKD-related hyperuricemia instead suppresses innate immunity.74 This phenomenon occurs through the intracellular uptake of soluble uric acid into CD14+ monocytes via selective urate transporters, such as the glucose transporter 9 (SLC2A9), which inhibits Toll-like receptor signaling, production of proinflammatory cytokines, and migration of CD14+ monocytes in patients with CKD/ESKD.74 Hyperuricemia also attenuates β2 integrin activation and integrin trafficking in neutrophils, which impairs neutrophil recruitment to sites of DAMP/PAMP-driven sterile inflammation (own unpublished data). This effect highlights a previously unexpected immunoregulatory role of asymptomatic serum UA levels of 7–12 mg/dl in those with CKD/ESKD. Further studies are needed to clarify the pathophysiologic effects of asymptomatic hyperuricemia in SIDKD, especially whether urate-lowering therapy can improve host defense in CKD/ESKD.
Immune Paralysis due to Increased Production of Immunoregulatory Proteins
A decline in kidney excretory function is associated with overexpression of certain immunoregulatory proteins (Figure 4). For example, fibroblast growth factor 23 (FGF23) levels drastically increase with CKD progression, which has a direct effect on neutrophil functions, including selectin-mediated slow rolling, chemokine-induced adhesion and arrest under dynamic flow, and ROS release. The resulting neutrophil recruitment defect impairs pathogen control in bacterial pneumonia in mice.71 Mechanistically, FGF23 binding to its counter-receptor FGFR2 on neutrophils counteracts selectin- and chemokine-triggered β2 integrin activation by activating protein kinase A and suppressing the activation of the small GTPase Rap1. Reduced neutrophil rolling and arrest on activated endothelium, associated with elevated FGF23 blood levels, was also observed in whole blood samples from patients with CKD stage G3–5.71
Levels of the middle-molecular-weight proteins leptin,160,161 resistin,161,162 and modified ubiquitin70 increase in patients with CKD/ESKD compared with healthy controls,162 which contributes to neutrophil dysfunction. Leptin can inhibit neutrophil migration by impairing neutrophil locomotion via MAPK- and Src kinase–related F-actin polymerization.160 In contrast, resistin162 and p-cresol163 decrease the respiratory burst activity of neutrophils, whereas complement factor D and angiogenin overexpression inhibit neutrophil degranulation164165–166 and glucose-modified proteins (glycated proteins) promote neutrophil apoptosis.167
SIDKD due to Extrinsic Immunosuppressors
Extrinsic factors contribute to SIDKD (Figure 4). For example, there is a concern that iron administration for renal anemia could decrease host defense. Indeed, iron can decrease the phagocytic capacity of neutrophils and macrophages.168,169 Iron can also impair the proliferation of T cells, lower IL-2 and IFN-γ production, and reduce the antibody response of B cells.170171–172 Infection may be a long-term complication of iron therapy in patients with CKD and may play an important role in suppressing immunity, especially in cases of iron overload.173 A meta-analysis of randomized controlled trials showed that intravenous iron therapy is associated with a 30% greater risk of infection compared with oral iron and no iron therapy.174 However, modern intravenous iron formulations do not seem to increase infection rate in patients on HD.175
Steroids and other immunosuppressant drugs are frequently prescribed to patients with CKD/ESKD and kidney transplant recipients for their anti-inflammatory and immunosuppressive effects. However, many clinical trials176177178179–180 (including the STOP-IgAN181 and TESTING182,183 trials) assessing the use of steroids, e.g., corticosteroids, prednisolone (initially 20–60 mg/d, then tapered, for 4–24 months), reported a high rate of serious adverse outcomes (STOP-IgAN, 55%; TESTING, risk difference, 11.5% [95% CI, 4.8% to 18.2%]), including an increased risk of infections and certain opportunistic infections (STOP-IgAN, 25%; TESTING, 8%). Another cohort study reported similar results, with a 40% higher risk of serious adverse events (46.1 per 1000 person-years; relative risk, 1.4 [95% CI, 1.3 to 1.6]) in patients with CKD treated with corticosteroids most of them infections184 Steroids are an independent risk factor for SID because they also increase the risk of infection in patients without CKD who have rheumatoid arthritis, asthma, vasculitis, or inflammatory bowel disease.185186187188–189 However, infectious complications are not increased in those receiving a daily dose <10 mg or a cumulative dose of >700 mg of prednisone.189 The extent to which SID is more severe in patients treated with steroids who have CKD versus those without CKD has not been established and will certainly also affect the many confounding factors contributing to SID. Mechanistically, these agents inhibit the release of proinflammatory cytokines (e.g., IL-1β, IL-6, TNFα) in monocytes/macrophages, the recruitment and respiratory burst in neutrophils, and cause lymphopenia and impair lymphocyte function.190191192–193 Although these drugs are sometimes needed to control the underlying kidney disease,194 balancing protective and dysregulated host immune responses can be intricate in patients who are critically ill and experience reinfection, e.g., with COVID-19.1,51
The removal of metabolic/uremic waste products by passing blood through synthetic tubing and dialyzer membranes during HD triggers the activation of monocytes, neutrophils, platelets, and the complement system; the release of inflammatory cytokines and ROS; and apoptosis.195,196 Neutrophil activation with decreased β2 integrin expression197 and neutrophil degranulation,198 along with the neutrophil extracellular trap formation associated with the release of DNA, myeloperoxidase, histones, and S100 proteins, can trigger endothelial activation,199 which ultimately contributes to vascular inflammation and cardiovascular disease.200 This may explain, at least in part, the cell-mediated immunosuppression and immune paralysis seen in patients with ESKD who are on HD.
Summary and a Call for Action
More awareness is needed for the unmet need of infection-related morbidity and mortality and the other clinical consequences of SID in patients with kidney disease. The ongoing pandemic provides a unique opportunity in this context, with kidney disease a major risk factor for lethal COVID-19 and a poor vaccine response. As a starting point, we provide a practical definition of SIDKD to endorse focused research on the epidemiology of this important cause of death in patients with kidney disease. As a community of kidney researchers, we have to improve patient outcomes in this domain as we do in CKD-related cardiovascular disease. To develop better preventive and therapeutic strategies, we first need to understand the pathophysiology of SIDKD (Table 3). Some mechanisms are discussed here but, undoubtedly, there are more to discover. SIDKD offers plenty of research opportunities for established and next-generation researchers, and opportunities for industry to invest in a naive domain for therapeutic interventions. The unmet needs of SIDKD require more attention at all levels.
Table 3. -
Approaches to improve SIDKD
Approaches to Identify and Prevent SIDKD |
Potential Diagnosis/Intervention |
Primary prevention of kidney disease |
Pregnancy counseling to avoid pregnancy complications, optimized nutrition of pregnant women, minimized exposure to nephrotoxins, healthy lifestyle, early diagnosis and treatment of systemic disorders (diabetes, amyloidosis, SLE) or infections (viral hepatitis, malaria, HIV, bacterial tonsillitis) |
Recognizing primary ID as a cause of kidney disease |
Clinical awareness, diagnosis, monitoring, and treatment of primary ID |
Secondary prevention of progression of kidney disease |
Disease-specific therapies; healthy lifestyle; smoking cessation; control of diabetes, BP, body weight, and metabolic acidosis; avoiding nephrotoxins and RAS/SGLT2 inhibitors; mineralocorticosteroid receptor antagonism (in diabetes); avoiding infections (vaccination) and rigorous treatment of infections |
Identifying predominant pathomechanisms of SIDKD |
Targeting specific pathomechanisms |
Clinical trials testing potential interventions using primary end points |
RAS, renin-angiotensin system; SGLT2, sodium-glucose transporter 2.
Disclosures
H.-J. Anders reports receiving consultancy or lecture fees from AstraZeneca, Bayer, Boehringer Ingelheim, Eleva, GlaxoSmithKline, Janssen, Kezar, Lilly, Novartis, Otsuka, and PreviPharma; and serving as a scientific advisor for, or member of, JASN and Nephrology Dialysis Transplantation. S. Steiger reports receiving research funding from Eleva. A. Zarbock reports serving as a scientific advisor for, or member of, AM Pharma, Anesthesia & Analgesia, Guard Therapeutics, Journal of Immunology, Novartis, and Piaon; and receiving consulting fees, unrestricted research grants, or lecture fees from AM Pharma, Anomed, Astellas, Astute Medical, Baxter, BiMerieux, Braun, Deutsche Forschungsgemeinschaft, Else-Kröner Fresenius Stiftung, Fresenius, Guard Therapeutics, La Jolla Pharmaceuticals, Novartis, and Ratiopharm. The remaining author has nothing to disclose.
Funding
S. Steiger was supported by the Deutsche Forschungsgemeinschaft grants STE2437/2-1 and STE2437/2-2 and the Ludwig-Maximilians-Universität München Excellence Initiative. H.-J. Anders was supported by the Deutsche Forschungsgemeinschaft grants AN372/14-4, 20-2, 27-1, and 30-1 and the Volkswagen Foundation grant 97-744. A. Zarbock was supported by the Deutsche Forschungsgemeinschaft grants ZA428/17-1, ZA428/18-1, and SFB1009A05. J. Rossaint was supported by the Deutsche Forschungsgemeinschaft grant RO4537/4-1 and the Interdisziplinäres Zentrum für Klinische Forschung, Universitätsklinikum Würzburg grant Ross2/010/18.
Acknowledgments
Because H.-J. Anders is an editor of JASN, he was not involved in the peer-review process for this manuscript. A guest editor oversaw the peer-review and decision-making process for this manuscript.
References
1. Kronbichler A, Gauckler P, Windpessl M, Il Shin J, Jha V, Rovin BH, et al.: COVID-19: Implications for immunosuppression in kidney disease and transplantation. Nat Rev Nephrol 16: 365–367, 2020
2. Seidel MG, Kindle G, Gathmann B, Quinti I, Buckland M, van Montfrans J, et al.; ESID Registry Working Party and collaborators: The European Society for Immunodeficiencies (ESID) Registry working definitions for the clinical diagnosis of inborn errors of immunity. J Allergy Clin Immunol Pract 7: 1763–1770, 2019
3. Monleón Bonet C, Waser N, Cheng K, Tzivelekis S, Edgar JDM, Sánchez-Ramón S: A systematic literature review of the effects of immunoglobulin replacement therapy on the burden of secondary immunodeficiency diseases associated with hematological malignancies and stem cell transplants. Expert Rev Clin Immunol 16: 911–921, 2020
4. Lano G, Braconnier A, Bataille S, Cavaille G, Moussi-Frances J, Gondouin B, et al.: Risk factors for severity of COVID-19 in chronic dialysis patients from a multicentre French cohort. Clin Kidney J 13: 878–888, 2020
5. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al.: Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395: 497–506, 2020
6. Grasselli G, Zangrillo A, Zanella A, Antonelli M, Cabrini L, Castelli A, et al.; COVID-19 Lombardy ICU Network: Baseline characteristics and outcomes of 1591 patients infected with SARS-CoV-2 admitted to ICUs of the Lombardy region, Italy. JAMA 323: 1574–1581, 2020
7. Alberici F, Delbarba E, Manenti C, Econimo L, Valerio F, Pola A, et al.: A report from the Brescia Renal COVID Task Force on the clinical characteristics and short-term outcome of hemodialysis patients with SARS-CoV-2 infection. Kidney Int 98: 20–26, 2020
8. Gibertoni D, Reno C, Rucci P, Fantini MP, Buscaroli A, Mosconi G, et al.: COVID-19 incidence and mortality in non-dialysis chronic kidney disease patients. PLoS One 16: e0254525, 2021
9. Goicoechea M, Sánchez Cámara LA, Macías N, Muñoz de Morales A, Rojas AG, Bascuñana A, et al.: COVID-19: Clinical course and outcomes of 36 hemodialysis patients in Spain. Kidney Int 98: 27–34, 2020
10. Broseta JJ, Rodríguez-Espinosa D, Cuadrado E, Guillén-Olmos E, Hermida E, Montagud-Marrahi E, et al.: SARS-CoV-2 infection in a Spanish cohort of CKD-5D patients: Prevalence, clinical presentation, outcomes, and de-isolation results. Blood Purif 50: 531–538, 2021
11. Kang SH, Kim SW, Kim AY, Cho KH, Park JW, Do JY: Association between chronic kidney disease or acute kidney injury and clinical outcomes in COVID-19 patients. J Korean Med Sci 35: e434, 2020
12. Ozturk S, Turgutalp K, Arici M, Gok M, Islam M, Altiparmak MR, et al.: Characteristics and outcomes of hospitalised older patients with chronic kidney disease and COVID-19: A multicenter nationwide controlled study. Int J Clin Pract 75: e14428, 2021
13. Pilgram L, Eberwein L, Wille K, Koehler FC, Stecher M, Rieg S, et al.; LEOSS Study group: Clinical course and predictive risk factors for fatal outcome of SARS-CoV-2 infection in patients with chronic kidney disease. Infection 49: 725–737, 2021
14. Akchurin O, Meza K, Biswas S, Greenbaum M, Licona-Freudenstein AP, Goyal P, et al.: COVID-19 in patients with CKD in New York City. Kidney360 2: 63–70, 2021
15. Navarrete JE, Tong DC, Cobb J, Rahbari-Oskoui FF, Hosein D, Caberto SC, et al.: Epidemiology of COVID-19 infection in hospitalized end-stage kidney disease patients in a predominantly African-American population. Am J Nephrol 52: 190–198, 2021
16. Smolander J, Bruchfeld A: The COVID-19 epidemic: Management and outcomes of hemodialysis and peritoneal dialysis patients in Stockholm, Sweden. Kidney Blood Press Res 46: 250–256, 2021
17. Chung EYM, Palmer SC, Natale P, Krishnan A, Cooper TE, Saglimbene VM, et al.: Incidence and outcomes of COVID-19 in people with CKD: A systematic review and meta-analysis. Am J Kidney Dis 78: 804–815, 2021
18. Clarke C, Lucisano G, Prendecki M, Gleeson S, Martin P, Ali M, et al.; ICHNT Renal COVID Group: Informing the risk of kidney transplantation versus remaining on the waitlist in the coronavirus disease 2019 era. Kidney Int Rep 6: 46–55, 2021
19. Heerspink HJL, Sjöström CD, Jongs N, Chertow GM, Kosiborod M, Hou FF, et al.; DAPA-CKD Trial Committees and Investigators: Effects of dapagliflozin on mortality in patients with chronic kidney disease: a pre-specified analysis from the DAPA-CKD randomized controlled trial. Eur Heart J 42: 1216–1227, 2021
20. Ishigami J, Grams ME, Chang AR, Carrero JJ, Coresh J, Matsushita K: CKD and risk for hospitalization with infection: The Atherosclerosis Risk in Communities (ARIC) Study. Am J Kidney Dis 69: 752–761, 2017
21. Meier-Kriesche HU, Ojo AO, Hanson JA, Kaplan B: Exponentially increased risk of infectious death in older renal transplant recipients. Kidney Int 59: 1539–1543, 2001
22. Vogelzang JL, van Stralen KJ, Noordzij M, Diez JA, Carrero JJ, Couchoud C, et al.: Mortality from infections and malignancies in patients treated with renal replacement therapy: Data from the ERA-EDTA registry. Nephrol Dial Transplant 30: 1028–1037, 2015
23. Saran R, Robinson B, Abbott KC, Agodoa LY, Albertus P, Ayanian J, et al.: US Renal Data System 2016 Annual Data Report: Epidemiology of kidney disease in the United States [published correction appears in
Am J Kidney Dis 69: 712, 2017]. Am J Kidney Dis 69: A7–A8, 2017
24. James MT, Laupland KB, Tonelli M, Manns BJ, Culleton BF, Hemmelgarn BR; Alberta Kidney Disease Network: Risk of bloodstream infection in patients with chronic kidney disease not treated with dialysis. Arch Intern Med 168: 2333–2339, 2008
25. Dalrymple LS, Katz R, Kestenbaum B, de Boer IH, Fried L, Sarnak MJ, et al.: The risk of infection-related hospitalization with decreased kidney function. Am J Kidney Dis 59: 356–363, 2012
26. James MT, Quan H, Tonelli M, Manns BJ, Faris P, Laupland KB, et al.; Alberta Kidney Disease Network: CKD and risk of hospitalization and death with pneumonia. Am J Kidney Dis 54: 24–32, 2009
27. Marcelli D, Marelli C, Richards N: Influenza A(H1N1)v pandemic in the dialysis population: First wave results from an international survey. Nephrol Dial Transplant 24: 3566–3572, 2009
28. Kwan BC, Leung CB, Szeto CC, Wong VW, Cheng YL, Yu AW, et al.: Severe acute respiratory syndrome in dialysis patients. J Am Soc Nephrol 15: 1883–1888, 2004
29. Wong PN, Mak SK, Lo KY, Tong GM, Wong Y, Watt CL, et al.: Clinical presentation and outcome of severe acute respiratory syndrome in dialysis patients. Am J Kidney Dis 42: 1075–1081, 2003
30. Ndamase S, Okpechi I, Carrara H, Black J, Calligaro G, Freercks R: Tuberculosis burden in stage 5 chronic kidney disease patients undergoing dialysis therapy at Livingstone Hospital, Port Elizabeth, South Africa. S Afr Med J 110: 422–426, 2020
31. Ruzangi J, Iwagami M, Smeeth L, Mangtani P, Nitsch D: The association between chronic kidney disease and tuberculosis; a comparative cohort study in England. BMC Nephrol 21: 420, 2020
32. Imtiaz S, Drohlia MF, Nasir K, Hussain M, Ahmad A: Morbidity and mortality associated with
Plasmodium vivax and
Plasmodium falciparum infection in a tertiary care kidney hospital. Saudi J Kidney Dis Transpl 26: 1169–1176, 2015
33. Simões-Silva L, Correia I, Barbosa J, Santos-Araujo C, Sousa MJ, Pestana M, et al.: Asymptomatic effluent protozoa colonization in peritoneal dialysis patients. Perit Dial Int 36: 566–569, 2016
34. Vanholder R, Ringoir S: Infectious morbidity and defects of phagocytic function in end-stage renal disease: A review. J Am Soc Nephrol 3: 1541–1554, 1993
35. Dalrymple LS, Go AS: Epidemiology of acute infections among patients with chronic kidney disease. Clin J Am Soc Nephrol 3: 1487–1493, 2008
36. Foley RN, Guo H, Snyder JJ, Gilbertson DT, Collins AJ: Septicemia in the United States dialysis population, 1991 to 1999. J Am Soc Nephrol 15: 1038–1045, 2004
37. Ludvigsen LU, Dalgaard LS, Wiggers H, Jensen-Fangel S, Jespersen B, Ellermann-Eriksen S, et al.: Infective endocarditis in patients receiving chronic hemodialysis: A 21-year observational cohort study in Denmark. Am Heart J 182: 36–43, 2016
38. Hoen B, Paul-Dauphin A, Hestin D, Kessler M: EPIBACDIAL: A multicenter prospective study of risk factors for bacteremia in chronic hemodialysis patients. J Am Soc Nephrol 9: 869–876, 1998
39. Ishani A, Collins AJ, Herzog CA, Foley RN: Septicemia, access and cardiovascular disease in dialysis patients: The USRDS Wave 2 study. Kidney Int 68: 311–318, 2005
40. Allon M, Depner TA, Radeva M, Bailey J, Beddhu S, Butterly D, et al.; HEMO Study Group: Impact of dialysis dose and membrane on infection-related hospitalization and death: Results of the HEMO Study. J Am Soc Nephrol 14: 1863–1870, 2003
41. Aslam N, Bernardini J, Fried L, Burr R, Piraino B: Comparison of infectious complications between incident hemodialysis and peritoneal dialysis patients. Clin J Am Soc Nephrol 1: 1226–1233, 2006
42. Li PK, Chow KM: Infectious complications in dialysis--epidemiology and outcomes. Nat Rev Nephrol 8: 77–88, 2011
43. Boyce JM, Dumigan DG, Havill NL, Hollis RJ, Pfaller MA, Moore BA: A multi-center outbreak of
Candida tropicalis bloodstream infections associated with contaminated hemodialysis machine prime buckets. Am J Infect Control 49: 1008–1013, 2021
44. Piccinelli G, De Francesco MA, Corbellini S, Lorenzin G, Caruso A: Detection of microbial contamination in dialysis water and gastrointestinal endoscopes by the Uro4 HB&L™ system. J Infect Public Health 13: 1054–1056, 2020
45. Strengert M, Becker M, Ramos GM, Dulovic A, Gruber J, Juengling J, et al.: Cellular and humoral immunogenicity of a SARS-CoV-2 mRNA vaccine in patients on haemodialysis. EBioMedicine 70: 103524, 2021
46. Garcia P, Anand S, Han J, Montez-Rath M, Sun S, Shang T, et al.: COVID19 vaccine type and humoral immune response in patients receiving dialysis. medRxiv. 2021.
47. Rincon-Arevalo H, Choi M, Stefanski AL, Halleck F, Weber U, Szelinski F, et al.: Impaired humoral immunity to SARS-CoV-2 BNT162b2 vaccine in kidney transplant recipients and dialysis patients. Sci Immunol 6: eabj1031, 2021
48. Sattler A, Schrezenmeier E, Weber UA, Potekhin A, Bachmann F, Straub-Hohenbleicher H, et al.: Impaired humoral and cellular immunity after SARS-CoV-2 BNT162b2 (tozinameran) prime-boost vaccination in kidney transplant recipients. J Clin Invest 131: 150175, 2021
49. Korth J, Jahn M, Dorsch O, Anastasiou OE, Sorge-Hädicke B, Eisenberger U, et al.: Impaired humoral response in renal transplant recipients to SARS-CoV-2 vaccination with BNT162b2 (Pfizer-BioNTech). Viruses 13: 756, 2021
50. Grupper A, Sharon N, Finn T, Cohen R, Israel M, Agbaria A, et al.: Humoral response to the Pfizer BNT162b2 vaccine in patients undergoing maintenance hemodialysis. Clin J Am Soc Nephrol 16: 1037–1042, 2021
51. Cohen DE, Sibbel S, Marlowe G, Bludorn K, Miller D, Kelley T, et al.: Antibody status, disease history, and incidence of SARS-CoV-2 infection among patients on chronic dialysis. J Am Soc Nephrol 32: 1880–1886, 2021
52. Speer C, Schaier M, Nusshag C, Töllner M, Buylaert M, Kälble F, et al.: Longitudinal humoral responses after COVID-19 vaccination in peritoneal and hemodialysis patients over twelve weeks. Vaccines (Basel) 9: 1130, 2021
53. Stumpf J, Siepmann T, Lindner T, Karger C, Schwöbel J, Anders L, et al.: Humoral and cellular immunity to SARS-CoV-2 vaccination in renal transplant versus dialysis patients: A prospective, multicenter observational study using mRNA-1273 or BNT162b2 mRNA vaccine. Lancet Reg Health Eur 9: 100178, 2021
54. Ferreira TMB, Guimarães TGS, Fontenele AMM, Salgado N, Ferreira ASP, Costa APM: Does infection by the hepatitis C virus decrease the response of immunization against the hepatitis B virus in individuals undergoing dialysis? J Bras Nefrol 39: 141–145, 2017
55. Agarwal SK, Irshad M, Dash SC: Comparison of two schedules of hepatitis B vaccination in patients with mild, moderate and severe renal failure. J Assoc Physicians India 47: 183–185, 1999
56. Peces R, de la Torre M, Alcázar R, Urra JM: Prospective analysis of the factors influencing the antibody response to hepatitis B vaccine in hemodialysis patients. Am J Kidney Dis 29: 239–245, 1997
57. Crosnier J, Jungers P, Couroucé AM, Laplanche A, Benhamou E, Degos F, et al.: Randomised placebo-controlled trial of hepatitis B surface antigen vaccine in French haemodialysis units: II, Haemodialysis patients. Lancet 1: 797–800, 1981
58. Liu YL, Kao MT, Huang CC: A comparison of responsiveness to hepatitis B vaccination in patients on hemodialysis and peritoneal dialysis. Vaccine 23: 3957–3960, 2005
59. Krüger S, Müller-Steinhardt M, Kirchner H, Kreft B: A 5-year follow-up on antibody response after diphtheria and tetanus vaccination in hemodialysis patients. Am J Kidney Dis 38: 1264–1270, 2001
60. Kreft B, Klouche M, Kreft R, Kirchner H, Sack K: Low efficiency of active immunization against diphtheria in chronic hemodialysis patients. Kidney Int 52: 212–216, 1997
61. Fuchshuber A, Kühnemund O, Keuth B, Lütticken R, Michalk D, Querfeld U: Pneumococcal vaccine in children and young adults with chronic renal disease. Nephrol Dial Transplant 11: 468–473, 1996
62. Beyer WE, Versluis DJ, Kramer P, Diderich PP, Weimar W, Masurel N: Trivalent influenza vaccine in patients on haemodialysis: Impaired seroresponse with differences for A-H3N2 and A-H1N1 vaccine components. Vaccine 5: 43–48, 1987
63. Ducloux D, Colladant M, Chabannes M, Yannaraki M, Courivaud C: Humoral response after 3 doses of the BNT162b2 mRNA COVID-19 vaccine in patients on hemodialysis. Kidney Int 100: 702–704, 2021
64. Juraschek SP, Kovell LC, Miller ER 3rd, Gelber AC: Association of kidney disease with prevalent gout in the United States in 1988–1994 and 2007–2010. Semin Arthritis Rheum 42: 551–561, 2013
65. Jing J, Kielstein JT, Schultheiss UT, Sitter T, Titze SI, Schaeffner ES, et al.; GCKD Study Investigators: Prevalence and correlates of gout in a large cohort of patients with chronic kidney disease: the German Chronic Kidney Disease (GCKD) study. Nephrol Dial Transplant 30: 613–621, 2015
66. Furman D, Campisi J, Verdin E, Carrera-Bastos P, Targ S, Franceschi C, et al.:
Chronic inflammation in the etiology of disease across the life span. Nat Med 25: 1822–1832, 2019
67. Morena M, Cristol JP, Senécal L, Leray-Moragues H, Krieter D, Canaud B: Oxidative stress in hemodialysis patients: Is NADPH oxidase complex the culprit? Kidney Int Suppl 61: 109–114, 2002
68. Rodríguez-Ayala E, Anderstam B, Suliman ME, Seeberger A, Heimbürger O, Lindholm B, et al.: Enhanced RAGE-mediated NFkappaB stimulation in inflamed hemodialysis patients. Atherosclerosis 180: 333–340, 2005
69. Ottonello L, Gnerre P, Bertolotto M, Mancini M, Dapino P, Russo R, et al.: Leptin as a uremic toxin interferes with neutrophil chemotaxis. J Am Soc Nephrol 15: 2366–2372, 2004
70. Cohen G, Rudnicki M, Hörl WH: Isolation of modified ubiquitin as a neutrophil chemotaxis inhibitor from uremic patients. J Am Soc Nephrol 9: 451–456, 1998
71. Rossaint J, Oehmichen J, Van Aken H, Reuter S, Pavenstädt HJ, Meersch M, et al.: FGF23 signaling impairs neutrophil recruitment and host defense during CKD. J Clin Invest 126: 962–974, 2016
72. Rossaint J, Spelten O, Kässens N, Mueller H, Van Aken HK, Singbartl K, et al.: Acute loss of renal function attenuates slow leukocyte rolling and transmigration by interfering with intracellular signaling. Kidney Int 80: 493–503, 2011
73. Zarbock A, Schmolke M, Spieker T, Jurk K, Van Aken H, Singbartl K: Acute uremia but not renal inflammation attenuates aseptic acute lung injury: A critical role for uremic neutrophils. J Am Soc Nephrol 17: 3124–3131, 2006
74. Ma Q, Honarpisheh M, Li C, Sellmayr M, Lindenmeyer M, Böhland C, et al.: Soluble uric acid is an intrinsic negative regulator of monocyte activation in monosodium urate crystal-induced tissue inflammation. J Immunol 205: 789–800, 2020
75. Glorieux G, Vanholder R, Lameire N: Uraemic retention and apoptosis: What is the balance for the inflammatory status in uraemia? Eur J Clin Invest 33: 631–634, 2003
76. Kato S, Chmielewski M, Honda H, Pecoits-Filho R, Matsuo S, Yuzawa Y, et al.: Aspects of immune dysfunction in end-stage renal disease. Clin J Am Soc Nephrol 3: 1526–1533, 2008
77. Anding K, Gross P, Rost JM, Allgaier D, Jacobs E: The influence of uraemia and haemodialysis on neutrophil phagocytosis and antimicrobial killing. Nephrol Dial Transplant 18: 2067–2073, 2003
78. Haag-Weber M, Cohen G, Hörl WH: Clinical significance of granulocyte-inhibiting proteins. Nephrol Dial Transplant 15[Suppl 1]: 15–16, 2000
79. Cohen G, Horl WH: Immune dysfunction in uremia—An update. Toxins (Basel) 4: 962–990, 2012
80. Himmelfarb J, Le P, Klenzak J, Freedman S, McMenamin ME, Ikizler TA; PICARD Group: Impaired monocyte cytokine production in critically ill patients with acute renal failure. Kidney Int 66: 2354–2360, 2004
81. Ando M, Shibuya A, Tsuchiya K, Akiba T, Nitta K: Reduced capacity of mononuclear cells to synthesize cytokines against an inflammatory stimulus in uremic patients. Nephron Clin Pract 104: c113–c119, 2006
82. Verkade MA, van Druningen CJ, Vaessen LM, Hesselink DA, Weimar W, Betjes MG: Functional impairment of monocyte-derived dendritic cells in patients with severe chronic kidney disease. Nephrol Dial Transplant 22: 128–138, 2007
83. Ando M, Shibuya A, Tsuchiya K, Akiba T, Nitta K: Reduced expression of Toll-like receptor 4 contributes to impaired cytokine response of monocytes in uremic patients. Kidney Int 70: 358–362, 2006
84. Girndt M, Sester M, Sester U, Kaul H, Köhler H: Defective expression of B7-2 (CD86) on monocytes of dialysis patients correlates to the uremia-associated immune defect. Kidney Int 59: 1382–1389, 2001
85. Satomura A, Endo M, Ohi H, Sudo S, Ohsawa I, Fujita T, et al.: Significant elevations in serum mannose-binding lectin levels in patients with chronic renal failure. Nephron 92: 702–704, 2002
86. Satomura A, Fujita T, Yanai M, Kumasaka K, Uehara Y, Okada K, et al.: Functional mannose-binding lectin levels in patients with end-stage renal disease on maintenance hemodialysis. J Innate Immun 4: 293–300, 2012
87. Ali YM, Ferrari M, Lynch NJ, Yaseen S, Dudler T, Gragerov S, et al.: Lectin pathway mediates complement activation by SARS-CoV-2 proteins. Front Immunol 12: 714511, 2021
88. Petersen SV, Thiel S, Jensenius JC: The mannan-binding lectin pathway of complement activation: Biology and disease association. Mol Immunol 38: 133–149, 2001
89. Jack DL, Klein NJ, Turner MW: Mannose-binding lectin: Targeting the microbial world for complement attack and opsonophagocytosis. Immunol Rev 180: 86–99, 2001
90. Noris M, Benigni A, Remuzzi G: The case of complement activation in COVID-19 multiorgan impact. Kidney Int 98: 314–322, 2020
91. Ando M, Lundkvist I, Bergström J, Lindholm B: Enhanced scavenger receptor expression in monocyte-macrophages in dialysis patients. Kidney Int 49: 773–780, 1996
92. Ando M, Gåfvels M, Bergström J, Lindholm B, Lundkvist I: Uremic serum enhances scavenger receptor expression and activity in the human monocytic cell line U937. Kidney Int 51: 785–792, 1997
93. Chmielewski M, Bryl E, Marzec L, Aleksandrowicz E, Witkowski JM, Rutkowski B: Expression of scavenger receptor CD36 in chronic renal failure patients. Artif Organs 29: 608–614, 2005
94. Schmidt S, Westhoff TH, Krauser P, Ignatius R, Jankowski J, Jankowski V, et al.: The uraemic toxin phenylacetic acid impairs macrophage function. Nephrol Dial Transplant 23: 3485–3493, 2008
95. Betjes MG, Tuk CW, Visser CE, Zemel D, Krediet RT, Arisz L, et al.: Analysis of the peritoneal cellular immune system during CAPD shortly before a clinical peritonitis. Nephrol Dial Transplant 9: 684–692, 1994
96. Sutherland TE, Shaw TN, Lennon R, Herrick SE, Rückerl D: Ongoing exposure to peritoneal dialysis fluid alters resident peritoneal macrophage phenotype and activation propensity. Front Immunol 12: 715209, 2021
97. Nagai K: Dysfunction of natural killer cells in end-stage kidney disease on hemodialysis. Ren Replace Ther 7: 8, 2021
98. Dounousi E, Duni A, Naka KK, Vartholomatos G, Zoccali C: The innate immune system and cardiovascular disease in ESKD: Monocytes and natural killer cells. Curr Vasc Pharmacol 19: 63–76, 2021
99. Rossaint J, Margraf A, Zarbock A: Role of platelets in leukocyte recruitment and resolution of inflammation. Front Immunol 9: 2712, 2018
100. Fryc J, Naumnik B: Thrombolome and its emerging role in chronic kidney diseases. Toxins (Basel) 13: 223, 2021
101. Florens N, Calzada C, Lemoine S, Boulet MM, Guillot N, Barba C, et al.: CKD increases carbonylation of HDL and is associated with impaired antiaggregant properties. J Am Soc Nephrol 31: 1462–1477, 2020
102. Schoorl M, Schoorl M, Nubé MJ, Bartels PC: Coagulation activation, depletion of platelet granules and endothelial integrity in case of uraemia and haemodialysis treatment. BMC Nephrol 14: 72, 2013
103. Sirolli V, Strizzi L, Di Stante S, Robuffo I, Procopio A, Bonomini M: Platelet activation and platelet-erythrocyte aggregates in end-stage renal disease patients on hemodialysis. Thromb Haemost 86: 834–839, 2001
104. Baaten CCFMJ, Sternkopf M, Henning T, Marx N, Jankowski J, Noels H: Platelet function in CKD: A systematic review and meta-analysis [published online ahead of print May 3, 2021]. J Am Soc Nephrol 10.1681/ASN.2020101440
105. Margraf A, Zarbock A: Platelets in inflammation and resolution. J Immunol 203: 2357–2367, 2019
106. Lutz PDMJ, Jurk PDRNK: Platelets in advanced chronic kidney disease: Two sides of the coin. Semin Thromb Hemost 46: 342–356, 2020
107. Eleftheriadis T, Antoniadi G, Liakopoulos V, Kartsios C, Stefanidis I: Disturbances of acquired immunity in hemodialysis patients. Semin Dial 20: 440–451, 2007
108. Yoon JW, Gollapudi S, Pahl MV, Vaziri ND: Naïve and central memory T-cell lymphopenia in end-stage renal disease. Kidney Int 70: 371–376, 2006
109. Moser B, Roth G, Brunner M, Lilaj T, Deicher R, Wolner E, et al.: Aberrant T cell activation and heightened apoptotic turnover in end-stage renal failure patients: A comparative evaluation between non-dialysis, haemodialysis, and peritoneal dialysis. Biochem Biophys Res Commun 308: 581–585, 2003
110. Meuer SC, Hauer M, Kurz P, Meyer zum Büschenfelde KH, Köhler H: Selective blockade of the antigen-receptor-mediated pathway of T cell activation in patients with impaired primary immune responses. J Clin Invest 80: 743–749, 1987
111. Stachowski J, Pollok M, Burrichter H, Spithaler C, Baldamus CA: Signalling via the TCR/CD3 antigen receptor complex in uremia is limited by the receptors number. Nephron 64: 369–375, 1993
112. Lisowska KA, Dębska-Ślizień A, Jasiulewicz A, Heleniak Z, Bryl E, Witkowski JM: Hemodialysis affects phenotype and proliferation of CD4-positive T lymphocytes. J Clin Immunol 32: 189–200, 2012
113. Xiaoyan J, Rongyi C, Xuesen C, Jianzhou Z, Jun J, Xiaoqiang D, et al.: The difference of T cell phenotypes in end stage renal disease patients under different dialysis modality. BMC Nephrol 20: 301, 2019
114. Ando M, Shibuya A, Yasuda M, Azuma N, Tsuchiya K, Akiba T, et al.: Impairment of innate cellular response to in vitro stimuli in patients on continuous ambulatory peritoneal dialysis. Nephrol Dial Transplant 20: 2497–2503, 2005
115. Sester U, Sester M, Hauk M, Kaul H, Köhler H, Girndt M: T-cell activation follows Th1 rather than Th2 pattern in haemodialysis patients. Nephrol Dial Transplant 15: 1217–1223, 2000
116. Vaziri ND, Pahl MV, Crum A, Norris K: Effect of uremia on structure and function of immune system. J Ren Nutr 22: 149–156, 2012
117. Fernández-Fresnedo G, Ramos MA, González-Pardo MC, de Francisco AL, López-Hoyos M, Arias M: B lymphopenia in uremia is related to an accelerated
in vitro apoptosis and dysregulation of Bcl-2. Nephrol Dial Transplant 15: 502–510, 2000
118. Jasiulewicz A, Lisowska KA, Dębska-Ślizień A, Witkowski JM: Phenotype, proliferation and apoptosis of B lymphocytes in hemodialysis patients treated with recombinant human erythropoietin. Int Immunol 28: 523–532, 2016
119. Molina M, Allende LM, Ramos LE, Gutiérrez E, Pleguezuelo DE, Hernández ER, et al.: CD19
+ B-cells, a new biomarker of mortality in hemodialysis patients. Front Immunol 9: 1221, 2018
120. Pahl MV, Gollapudi S, Sepassi L, Gollapudi P, Elahimehr R, Vaziri ND: Effect of end-stage renal disease on B-lymphocyte subpopulations, IL-7, BAFF and BAFF receptor expression. Nephrol Dial Transplant 25: 205–212, 2010
121. Rautenberg P, Teifke I, Schlegelberger T, Ullmann U: Influenza subtype-specific IgA, IgM and IgG responses in patients on hemodialysis after influenza vaccination. Infection 16: 323–328, 1988
122. Stevens CE, Alter HJ, Taylor PE, Zang EA, Harley EJ, Szmuness W: Hepatitis B vaccine in patients receiving hemodialysis. Immunogenicity and efficacy. N Engl J Med 311: 496–501, 1984
123. McIntyre CW, Harrison LE, Eldehni MT, Jefferies HJ, Szeto CC, John SG, et al.: Circulating endotoxemia: A novel factor in systemic inflammation and cardiovascular disease in chronic kidney disease. Clin J Am Soc Nephrol 6: 133–141, 2011
124. Bossola M, Di Stasio E, Sanguinetti M, Posteraro B, Antocicco M, Pepe G, et al.: Serum endotoxin activity measured with endotoxin activity assay is associated with serum interleukin-6 levels in patients on chronic hemodialysis. Blood Purif 42: 294–300, 2016
125. Wang F, Zhang P, Jiang H, Cheng S: Gut bacterial translocation contributes to microinflammation in experimental uremia. Dig Dis Sci 57: 2856–2862, 2012
126. Meijers B, Evenepoel P, Anders HJ: Intestinal microbiome and fitness in kidney disease. Nat Rev Nephrol 15: 531–545, 2019
127. Andersen K, Kesper MS, Marschner JA, Konrad L, Ryu M, Kumar Vr S, et al.: Intestinal dysbiosis, barrier dysfunction, and bacterial translocation account for CKD-related systemic inflammation. J Am Soc Nephrol 28: 76–83, 2017
128. Anders HJ, Andersen K, Stecher B: The intestinal microbiota, a leaky gut, and abnormal immunity in kidney disease. Kidney Int 83: 1010–1016, 2013
129. Zhang Z, Zhang G, Guo M, Tao W, Liu X, Wei H, et al.: The potential role of an aberrant mucosal immune response to SARS-CoV-2 in the pathogenesis of IgA nephropathy. Pathogens 10: 881, 2021
130. Biswas SK, Lopez-Collazo E: Endotoxin tolerance: New mechanisms, molecules and clinical significance. Trends Immunol 30: 475–487, 2009
131. López-Collazo E, del Fresno C: Pathophysiology of endotoxin tolerance: Mechanisms and clinical consequences. Crit Care 17: 242, 2013
132. Hotchkiss RS, Coopersmith CM, McDunn JE, Ferguson TA: The sepsis seesaw: Tilting toward immunosuppression. Nat Med 15: 496–497, 2009
133. Draisma A, Pickkers P, Bouw MP, van der Hoeven JG: Development of endotoxin tolerance in humans i
n vivo. Crit Care Med 37: 1261–1267, 2009
134. Ni Choileain N, MacConmara M, Zang Y, Murphy TJ, Mannick JA, Lederer JA: Enhanced regulatory T cell activity is an element of the host response to injury. J Immunol 176: 225–236, 2006
135. Delano MJ, Scumpia PO, Weinstein JS, Coco D, Nagaraj S, Kelly-Scumpia KM, et al.: MyD88-dependent expansion of an immature GR-1(+)CD11b(+) population induces T cell suppression and Th2 polarization in sepsis. J Exp Med 204: 1463–1474, 2007
136. Limaye AP, Kirby KA, Rubenfeld GD, Leisenring WM, Bulger EM, Neff MJ, et al.: Cytomegalovirus reactivation in critically ill immunocompetent patients. JAMA 300: 413–422, 2008
137. Luyt CE, Combes A, Deback C, Aubriot-Lorton MH, Nieszkowska A, Trouillet JL, et al.:
Herpes simplex virus lung infection in patients undergoing prolonged mechanical ventilation. Am J Respir Crit Care Med 175: 935–942, 2007
138. del Fresno C, Gómez-Piña V, Lores V, Soares-Schanoski A, Fernández-Ruiz I, Rojo B, et al.: Monocytes from cystic fibrosis patients are locked in an LPS tolerance state: Down-regulation of TREM-1 as putative underlying mechanism. PLoS One 3: e2667, 2008
139. del Fresno C, Soler-Rangel L, Soares-Schanoski A, Gómez-Piña V, González-León MC, Gómez-García L, et al.: Inflammatory responses associated with acute coronary syndrome up-regulate IRAK-M and induce endotoxin tolerance in circulating monocytes. J Endotoxin Res 13: 39–52, 2007
140. del Fresno C, García-Rio F, Gómez-Piña V, Soares-Schanoski A, Fernández-Ruíz I, Jurado T, et al.: Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients [published correction appears in
J Immunol 193: 2194, 2009]. J Immunol 182: 6494–6507, 2009
141. Biswas SK, Gangi L, Paul S, Schioppa T, Saccani A, Sironi M, et al.: A distinct and unique transcriptional program expressed by tumor-associated macrophages (defective NF-kappaB and enhanced IRF-3/STAT1 activation). Blood 107: 2112–2122, 2006
142. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al.: Diet rapidly and reproducibly alters the human gut microbiome. Nature 505: 559–563, 2014
143. Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, et al.: Host-gut microbiota metabolic interactions. Science 336: 1262–1267, 2012
144. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI: An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444: 1027–1031, 2006
145. Vaziri ND, Wong J, Pahl M, Piceno YM, Yuan J, DeSantis TZ, et al.: Chronic kidney disease alters intestinal microbial flora. Kidney Int 83: 308–315, 2013
146. Mafra D, Barros AF, Fouque D: Dietary protein metabolism by gut microbiota and its consequences for chronic kidney disease patients. Future Microbiol 8: 1317–1323, 2013
147. Lau WL, Savoj J, Nakata MB, Vaziri ND: Altered microbiome in chronic kidney disease: systemic effects of gut-derived uremic toxins. Clin Sci (Lond) 132: 509–522, 2018
148. Huang W, Zhou L, Guo H, Xu Y, Xu Y: The role of short-chain fatty acids in kidney injury induced by gut-derived inflammatory response. Metabolism 68: 20–30, 2017
149. Rossi M, Campbell KL, Johnson DW, Stanton T, Vesey DA, Coombes JS, et al.: Protein-bound uremic toxins, inflammation and oxidative stress: a cross-sectional study in stage 3-4 chronic kidney disease. Arch Med Res 45: 309–317, 2014
150. Ahmed MS, Abed M, Voelkl J, Lang F: Triggering of suicidal erythrocyte death by uremic toxin indoxyl sulfate. BMC Nephrol 14: 244, 2013
151. Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bartolomaeus H, et al.: Salt-responsive gut commensal modulates T
H17 axis and disease. Nature 551: 585–589, 2017
152. Poesen R, Windey K, Neven E, Kuypers D, De Preter V, Augustijns P, et al.: The influence of CKD on colonic microbial metabolism. J Am Soc Nephrol 27: 1389–1399, 2016
153. Xu KY, Xia GH, Lu JQ, Chen MX, Zhen X, Wang S, et al.: Impaired renal function and dysbiosis of gut microbiota contribute to increased trimethylamine-N-oxide in chronic kidney disease patients. Sci Rep 7: 1445, 2017
154. Azevedo ML, Bonan NB, Dias G, Brehm F, Steiner TM, Souza WM, et al.: p-Cresyl sulfate affects the oxidative burst, phagocytosis process, and antigen presentation of monocyte-derived macrophages. Toxicol Lett 263: 1–5, 2016
155. Yu M, Kim YJ, Kang DH: Indoxyl sulfate-induced endothelial dysfunction in patients with chronic kidney disease via an induction of oxidative stress. Clin J Am Soc Nephrol 6: 30–39, 2011
156. Ito S, Higuchi Y, Yagi Y, Nishijima F, Yamato H, Ishii H, et al.: Reduction of indoxyl sulfate by AST-120 attenuates monocyte inflammation related to chronic kidney disease. J Leukoc Biol 93: 837–845, 2013
157. Macpherson ME, Hov JR, Ueland T, Dahl TB, Kummen M, Otterdal K, et al.: Gut microbiota-dependent trimethylamine N-oxide associates with inflammation in common variable immunodeficiency. Front Immunol 11: 574500, 2020
158. Resnick ES, Moshier EL, Godbold JH, Cunningham-Rundles C: Morbidity and mortality in common variable immune deficiency over 4 decades. Blood 119: 1650–1657, 2012
159. Jørgensen SF, Fevang B, Aukrust P: Autoimmunity and inflammation in CVID: A possible crosstalk between immune activation, gut microbiota, and epigenetic modifications. J Clin Immunol 39: 30–36, 2019
160. Montecucco F, Bianchi G, Gnerre P, Bertolotto M, Dallegri F, Ottonello L: Induction of neutrophil chemotaxis by leptin: Crucial role for p38 and Src kinases. Ann N Y Acad Sci 1069: 463–471, 2006
161. Díez JJ, Iglesias P, Fernández-Reyes MJ, Aguilera A, Bajo MA, Alvarez-Fidalgo P, et al.: Serum concentrations of leptin, adiponectin and resistin, and their relationship with cardiovascular disease in patients with end-stage renal disease. Clin Endocrinol (Oxf) 62: 242–249, 2005
162. Cohen G, Ilic D, Raupachova J, Hörl WH: Resistin inhibits essential functions of polymorphonuclear leukocytes. J Immunol 181: 3761–3768, 2008
163. Vanholder R, De Smet R, Waterloos MA, Van Landschoot N, Vogeleere P, Hoste E, et al.: Mechanisms of uremic inhibition of phagocyte reactive species production: characterization of the role of p-cresol. Kidney Int 47: 510–517, 1995
164. Balke N, Holtkamp U, Hörl WH, Tschesche H: Inhibition of degranulation of human polymorphonuclear leukocytes by complement factor D. FEBS Lett 371: 300–302, 1995
165. Tschesche H, Kopp C, Hörl WH, Hempelmann U: Inhibition of degranulation of polymorphonuclear leukocytes by angiogenin and its tryptic fragment. J Biol Chem 269: 30274–30280, 1994
166. Schmaldienst S, Oberpichler A, Tschesche H, Hörl WH: Angiogenin: A novel inhibitor of neutrophil lactoferrin release during extracorporeal circulation. Kidney Blood Press Res 26: 107–112, 2003
167. Cohen G, Rudnicki M, Walter F, Niwa T, Hörl WH: Glucose-modified proteins modulate essential functions and apoptosis of polymorphonuclear leukocytes. J Am Soc Nephrol 12: 1264–1271, 2001
168. Patruta SI, Edlinger R, Sunder-Plassmann G, Hörl WH: Neutrophil impairment associated with iron therapy in hemodialysis patients with functional iron deficiency. J Am Soc Nephrol 9: 655–663, 1998
169. Otaki Y, Nakanishi T, Hasuike Y, Moriguchi R, Nanami M, Hama Y, et al.: Defective regulation of iron transporters leading to iron excess in the polymorphonuclear leukocytes of patients on maintenance hemodialysis. Am J Kidney Dis 43: 1030–1039, 2004
170. Patruta SI, Hörl WH: Iron and infection. Kidney Int Suppl 69: S125–S130, 1999
171. Zhang Y, Wu L, Yang J, Zhou C, Liu Y: A nomogram-based prediction for severe pneumonia in patients with coronavirus disease 2019 (COVID-19). Infect Drug Resist 13: 3575–3582, 2020
172. Shaw J, Chakraborty A, Nag A, Chattopadyay A, Dasgupta AK, Bhattacharyya M: Intracellular iron overload leading to DNA damage of lymphocytes and immune dysfunction in thalassemia major patients. Eur J Haematol 99: 399–408, 2017
173. Sengoelge G, Sunder-Plassmann G, Hörl WH: Potential risk for infection and atherosclerosis due to iron therapy. J Ren Nutr 15: 105–110, 2005
174. Brookhart MA, Freburger JK, Ellis AR, Wang L, Winkelmayer WC, Kshirsagar AV: Infection risk with bolus versus maintenance iron supplementation in hemodialysis patients. J Am Soc Nephrol 24: 1151–1158, 2013
175. Macdougall IC, White C, Anker SD, Bhandari S, Farrington K, Kalra PA, et al.; PIVOTAL Investigators and Committees: Intravenous iron in patients undergoing maintenance hemodialysis [published correction appears in
N Engl J Med 380: 502, 2019]. N Engl J Med 380: 447–458, 2019
176. Lai KN, Lai FM, Ho CP, Chan KW: Corticosteroid therapy in IgA nephropathy with nephrotic syndrome: a long-term controlled trial. Clin Nephrol 26: 174–180, 1986
177. Koike M, Takei T, Uchida K, Honda K, Moriyama T, Horita S, et al.: Clinical assessment of low-dose steroid therapy for patients with IgA nephropathy: A prospective study in a single center. Clin Exp Nephrol 12: 250–255, 2008
178. Manno C, Torres DD, Rossini M, Pesce F, Schena FP: Randomized controlled clinical trial of corticosteroids plus ACE-inhibitors with long-term follow-up in proteinuric IgA nephropathy. Nephrol Dial Transplant 24: 3694–3701, 2009
179. Hogg RJ, Lee J, Nardelli N, Julian BA, Cattran D, Waldo B, et al.; Southwest Pediatric Nephrology Study Group: Clinical trial to evaluate omega-3 fatty acids and alternate day prednisone in patients with IgA nephropathy: Report from the Southwest Pediatric Nephrology Study Group. Clin J Am Soc Nephrol 1: 467–474, 2006
180. Katafuchi R, Ikeda K, Mizumasa T, Tanaka H, Ando T, Yanase T, et al.: Controlled, prospective trial of steroid treatment in IgA nephropathy: A limitation of low-dose prednisolone therapy. Am J Kidney Dis 41: 972–983, 2003
181. Rauen T, Eitner F, Fitzner C, Sommerer C, Zeier M, Otte B, et al.; STOP-IgAN Investigators: Intensive supportive care plus immunosuppression in IgA nephropathy. N Engl J Med 373: 2225–2236, 2015
182. Lv J, Zhang H, Wong MG, Jardine MJ, Hladunewich M, Jha V, et al.; TESTING Study Group: Effect of oral methylprednisolone on clinical outcomes in patients with IgA nephropathy: The TESTING Randomized Clinical Trial. JAMA 318: 432–442, 2017
183. Lv J, Xu D, Perkovic V, Ma X, Johnson DW, Woodward M, et al.; TESTING Study Group: Corticosteroid therapy in IgA nephropathy. J Am Soc Nephrol 23: 1108–1116, 2012
184. Oh GJ, Waldo A, Paez-Cruz F, Gipson PE, Pesenson A, Selewski DT, et al.: Steroid-associated side effects in patients with primary proteinuric kidney disease. Kidney Int Rep 4: 1608–1616, 2019
185. Fardet L, Petersen I, Nazareth I: Common infections in patients prescribed systemic glucocorticoids in primary care: A population-based cohort study. PLoS Med 13: e1002024, 2016
186. Chaudhary NS, Donnelly JP, Moore JX, Baddley JW, Safford MM, Wang HE: Association of baseline steroid use with long-term rates of infection and sepsis in the REGARDS cohort. Crit Care 21: 185, 2017
187. Dixon WG, Abrahamowicz M, Beauchamp ME, Ray DW, Bernatsky S, Suissa S, et al.: Immediate and delayed impact of oral glucocorticoid therapy on risk of serious infection in older patients with rheumatoid arthritis: A nested case-control analysis. Ann Rheum Dis 71: 1128–1133, 2012
188. Hoes JN, Jacobs JW, Verstappen SM, Bijlsma JW, Van der Heijden GJ: Adverse events of low- to medium-dose oral glucocorticoids in inflammatory diseases: A meta-analysis. Ann Rheum Dis 68: 1833–1838, 2009
189. Stuck AE, Minder CE, Frey FJ: Risk of infectious complications in patients taking glucocorticosteroids. Rev Infect Dis 11: 954–963, 1989
190. McEwen BS, Biron CA, Brunson KW, Bulloch K, Chambers WH, Dhabhar FS, et al.: The role of adrenocorticoids as modulators of immune function in health and disease: Neural, endocrine and immune interactions. Brain Res Brain Res Rev 23: 79–133, 1997
191. Boumpas DT, Chrousos GP, Wilder RL, Cupps TR, Balow JE: Glucocorticoid therapy for immune-mediated diseases: Basic and clinical correlates. Ann Intern Med 119: 1198–1208, 1993
192. Coutinho AE, Chapman KE: The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Mol Cell Endocrinol 335: 2–13, 2011
193. Youssef J, Novosad SA, Winthrop KL: Infection risk and safety of corticosteroid use. Rheum Dis Clin North Am 42: 157–176, ix–x, 2016
194. Karuthu S, Blumberg EA: Common infections in kidney transplant recipients. Clin J Am Soc Nephrol 7: 2058–2070, 2012
195. Himmelfarb J, Lazarus JM, Hakim R: Reactive oxygen species production by monocytes and polymorphonuclear leukocytes during dialysis. Am J Kidney Dis 17: 271–276, 1991
196. Hörl WH: Hemodialysis membranes: Interleukins, biocompatibility, and middle molecules. J Am Soc Nephrol 13: S62–S71, 2002
197. Lawrence MB, Springer TA: Leukocytes roll on a selectin at physiologic flow rates: Distinction from and prerequisite for adhesion through integrins. Cell 65: 859–873, 1991
198. Hörl WH, Steinhauer HB, Schollmeyer P: Plasma levels of granulocyte elastase during hemodialysis: Effects of different dialyzer membranes. Kidney Int 28: 791–796, 1985
199. Bieber S, Muczynski KA, Lood C: Neutrophil activation and neutrophil extracellular trap formation in dialysis patients. Kidney Med 2: 692–698.e1, 2020
200. Ekdahl KN, Soveri I, Hilborn J, Fellström B, Nilsson B: Cardiovascular disease in haemodialysis: Role of the intravascular innate immune system. Nat Rev Nephrol 13: 285–296, 2017
201. Gierski F, Stefaniak N, Benzerouk F, Gobin P, Schmid F, Henry A, et al.: Component process analysis of verbal memory in a sample of students with a binge drinking pattern. Addict Behav Rep 12: 100323, 2020
202. Xu H, Gasparini A, Ishigami J, Mzayen K, Su G, Barany P, et al.: eGFR and the risk of community-acquired infections. Clin J Am Soc Nephrol 12: 1399–1408, 2017
203. Naqvi SB, Collins AJ: Infectious complications in chronic kidney disease. Adv Chronic Kidney Dis 13: 199–204, 2006
204. Cheikh Hassan HI, Tang M, Djurdjev O, Langsford D, Sood MM, Levin A: Infection in advanced chronic kidney disease leads to increased risk of cardiovascular events, end-stage kidney disease and mortality. Kidney Int 90: 897–904, 2016
205. Lafrance JP, Rahme E, Iqbal S, Elftouh N, Vallée M, Laurin LP, et al.: Association of dialysis modality with risk for infection-related hospitalization: a propensity score-matched cohort analysis. Clin J Am Soc Nephrol 7: 1598–1605, 2012
206. Johnson DW, Dent H, Hawley CM, McDonald SP, Rosman JB, Brown FG, et al.: Associations of dialysis modality and infectious mortality in incident dialysis patients in Australia and New Zealand. Am J Kidney Dis 53: 290–297, 2009
207. Leon-Abarca JA, Memon RS, Rehan B, Iftikhar M, Chatterjee A: The impact of COVID-19 in diabetic kidney disease and chronic kidney disease: A population-based study. Acta Biomed 91: e2020161, 2020
208. Sran K, Olsburgh J, Kasimatis T, Clark K, Gökmen R, Hilton R, et al.: COVID-19 in kidney transplant patients from a large UK transplant center: Exploring risk factors for disease severity. Transplant Proc 53: 1160–1168, 2021
209. Goffin E, Candellier A, Vart P, Noordzij M, Arnol M, Covic A, et al.; ERACODA Collaborators: COVID-19-related mortality in kidney transplant and haemodialysis patients: A comparative, prospective registry-based study. Nephrol Dial Transplant 36: 2094–2105, 2021
210. Chen CY, Shao SC, Chen YT, Hsu CK, Hsu HJ, Lee CC, et al.: Incidence and clinical impacts of COVID-19 infection in patients with hemodialysis: Systematic review and meta-analysis of 396,062 hemodialysis patients. Healthcare (Basel) 9: 47, 2021
211. Hsu CM, Weiner DE, Aweh G, Miskulin DC, Manley HJ, Stewart C, et al.: COVID-19 among US dialysis patients: Risk factors and outcomes from a national dialysis provider. Am J Kidney Dis 77: 748–756.e1, 2021
212. Turgutalp K, Ozturk S, Arici M, Eren N, Gorgulu N, Islam M, et al.: Determinants of mortality in a large group of hemodialysis patients hospitalized for COVID-19. BMC Nephrol 22: 29, 2021
213. Moran E, Baharani J, Dedicoat M, Robinson E, Smith G, Bhomra P, et al.: Risk factors associated with the development of active tuberculosis among patients with advanced chronic kidney disease. J Infect 77: 291–295, 2018
214. Chia S, Karim M, Elwood RK, FitzGerald JM: Risk of tuberculosis in dialysis patients: A population-based study. Int J Tuberc Lung Dis 2: 989–991, 1998
215. Al-Efraij K, Mota L, Lunny C, Schachter M, Cook V, Johnston J: Risk of active tuberculosis in chronic kidney disease: A systematic review and meta-analysis. Int J Tuberc Lung Dis 19: 1493–1499, 2015
216. Igari H, Imasawa T, Noguchi N, Nagayoshi M, Mizuno S, Ishikawa S, et al.: Advanced stage of chronic kidney disease is risk of poor treatment outcome for smear-positive pulmonary tuberculosis. J Infect Chemother 21: 559–563, 2015
217. Cheng KC, Liao KF, Lin CL, Liu CS, Lai SW: Chronic kidney disease correlates with increased risk of pulmonary tuberculosis before initiating renal replacement therapy: A cohort study in Taiwan. Medicine (Baltimore) 97: e12550, 2018
218. Wu MY, Hsu YH, Su CL, Lin YF, Lin HW: Risk of herpes zoster in CKD: A matched-cohort study based on administrative data. Am J Kidney Dis 60: 548–552, 2012
219. Kim SC, Seo MY, Lee JY, Kim KT, Cho E, Kim MG, et al.: Advanced chronic kidney disease: A strong risk factor for
Clostridium difficile infection. Korean J Intern Med (Korean Assoc Intern Med) 31: 125–133, 2016
220. Eddi R, Malik MN, Shakov R, Baddoura WJ, Chandran C, Debari VA: Chronic kidney disease as a risk factor for
Clostridium difficile infection. Nephrology (Carlton) 15: 471–475, 2010
221. Morfin-Otero R, Garza-Gonzalez E, Garcia Garcia G, Perez-Gomez HR, Aguirre-Diaz SA, Gonzalez-Diaz E, et al.:
Clostridium difficile infection in patients with chronic kidney disease in Mexico. Clin Nephrol 90: 350–356, 2018
222. Lis Ł, Jerzak P, Konieczny A, Sroka M, Noceń-Rychlewska B, Podgórski P, et al.: Risk factors of the
Clostridium difficile infection in patients with chronic kidney disease. Adv Clin Exp Med 27: 1081–1084, 2018
223. Eui Oh S, Lee SM, Lee YK, Choi SR, Choi MJ, Kim JK, et al.:
Clostridium difficile-associated diarrhea in dialysis patients. Kidney Res Clin Pract 32: 27–31, 2013
224. McDonald HI, Thomas SL, Millett ER, Nitsch D: CKD and the risk of acute, community-acquired infections among older people with diabetes mellitus: A retrospective cohort study using electronic health records. Am J Kidney Dis 66: 60–68, 2015
225. Chou CY, Wang SM, Liang CC, Chang CT, Liu JH, Wang IK, et al.: Risk of pneumonia among patients with chronic kidney disease in outpatient and inpatient settings: A nationwide population-based study. Medicine (Baltimore) 93: e174, 2014
226. Viasus D, Garcia-Vidal C, Cruzado JM, Adamuz J, Verdaguer R, Manresa F, et al.: Epidemiology, clinical features and outcomes of pneumonia in patients with chronic kidney disease. Nephrol Dial Transplant 26: 2899–2906, 2011
227. Chen CH, Hsu WH, Chen HJ, Chen W, Shih CM, Hsia TC, et al.: Different bacteriology and prognosis of thoracic empyemas between patients with chronic and end-stage renal disease. Chest 132: 532–539, 2007
228. Maizel J, Deransy R, Dehedin B, Secq E, Zogheib E, Lewandowski E, et al.: Impact of non-dialysis chronic kidney disease on survival in patients with septic shock. BMC Nephrol 14: 77, 2013
229. Mansur A, Mulwande E, Steinau M, Bergmann I, Popov AF, Ghadimi M, et al.: Chronic kidney disease is associated with a higher 90-day mortality than other chronic medical conditions in patients with sepsis. Sci Rep 5: 10539, 2015
230. Weinhandl ED, Gilbertson DT, Collins AJ: Mortality, hospitalization, and technique failure in daily home hemodialysis and matched peritoneal dialysis patients: A matched cohort study. Am J Kidney Dis 67: 98–110, 2016
231. Sahli F, Feidjel R, Laalaoui R: Hemodialysis catheter-related infection: Rates, risk factors and pathogens. J Infect Public Health 10: 403–408, 2017
232. Ravani P, Palmer SC, Oliver MJ, Quinn RR, MacRae JM, Tai DJ, et al.: Associations between hemodialysis access type and clinical outcomes: A systematic review. J Am Soc Nephrol 24: 465–473, 2013
233. Delistefani F, Wallbach M, Müller GA, Koziolek MJ, Grupp C: Risk factors for catheter-related infections in patients receiving permanent dialysis catheter. BMC Nephrol 20: 199, 2019
234. Mangram AJ, Archibald LK, Hupert M, Tokars JI, Silver LC, Brennan P, et al.: Outbreak of sterile peritonitis among continuous cycling peritoneal dialysis patients. Kidney Int 54: 1367–1371, 1998
235. Lyman M, Nguyen DB, Shugart A, Gruhler H, Lines C, Patel PR: Risk of vascular access infection associated with buttonhole cannulation of fistulas: Data from the National Healthcare Safety Network. Am J Kidney Dis 76: 82–89, 2020
236. Golper TA, Brier ME, Bunke M, Schreiber MJ, Bartlett DK, Hamilton RW, et al.: Risk factors for peritonitis in long-term peritoneal dialysis: The Network 9 peritonitis and catheter survival studies. Academic Subcommittee of the Steering Committee of the Network 9 Peritonitis and Catheter Survival Studies. Am J Kidney Dis 28: 428–436, 1996
237. Eknoyan G, Beck GJ, Cheung AK, Daugirdas JT, Greene T, Kusek JW, et al.; Hemodialysis (HEMO) Study Group: Effect of dialysis dose and membrane flux in maintenance hemodialysis. N Engl J Med 347: 2010–2019, 2002
238. Doulton T, Sabharwal N, Cairns HS, Schelenz S, Eykyn S, O’Donnell P, et al.: Infective endocarditis in dialysis patients: new challenges and old. Kidney Int 64: 720–727, 2003
239. Zacharioudakis IM, Zervou FN, Ziakas PD, Mylonakis E: Meta-analysis of methicillin-resistant
Staphylococcus aureus colonization and risk of infection in dialysis patients. J Am Soc Nephrol 25: 2131–2141, 2014
240. Chaudry MS, Gislason GH, Kamper AL, Rix M, Larsen AR, Petersen A, et al.: Increased risk of
Staphylococcus aureus bacteremia in hemodialysis-A nationwide study. Hemodial Int 23: 230–238, 2019
241. Gulcan A, Gulcan E, Keles M, Aktas E: Oral yeast colonization in peritoneal dialysis and hemodialysis patients and renal transplant recipients. Comp Immunol Microbiol Infect Dis 46: 47–52, 2016
242. Li ZY, Chen M, Zhao MH: Severe infections following rituximab treatment in antineutrophil cytoplasmic antibody-associated vasculitis. Kidney Dis 7: 50–56, 2021