Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Dec 30;15(1):10842.
doi: 10.1038/s41467-024-55187-w.

The survival of B cells is compromised in kidney disease

Affiliations

The survival of B cells is compromised in kidney disease

Doureradjou Peroumal et al. Nat Commun. .

Abstract

Antibody-mediated protection against pathogens is crucial to a healthy life. However, the recent SARS-CoV-2 pandemic has shown that pre-existing comorbid conditions including kidney disease account for compromised humoral immunity to infections. Individuals with kidney disease are not only susceptible to infections but also exhibit poor vaccine-induced antibody response. Using multiple mouse models of kidney disease, we demonstrate that renal dysfunction inhibits germinal center (GC) response against T-dependent antigens. GC B cells exhibit increased apoptosis in kidney disease. Uremic toxin hippuric acid drives loss of mitochondrial membrane potential, leading to increased apoptosis of GC B cells in a G-protein-coupled receptor 109A dependent manner. Finally, GC B cells and antibody titer are diminished in mice with kidney disease following influenza virus infection, a major cause of mortality in individuals with renal disorders. These results provide a mechanistic understanding of how renal dysfunction suppresses humoral immunity in patients with kidney disease.

PubMed Disclaimer

Conflict of interest statement

Competing interests: The authors declare no competing interests.

Figures

Fig. 1
Fig. 1. Impaired GC formation in AAN mice.
A Schematic diagram of the experimental design. C57BL/6 (WT) mice were i.p. injected with a single dose of AAI, AAII (7.5 mg/kg b.wt), or PBS (Ctrl). Mice were either immunized with NP-KLH in alum or left non-immunized 4 days post-AAI injection. At 12 days post-immunization, spleens were assessed for B B cells (liveB220+) [Non-immun: Ctrl (3), AAI (3), AAII (3); NP-KLH: Ctrl (9), AAI (9), AAII (9)] C total GC B cells (liveB220+GL7+CD95+) [Non-immun: Ctrl (3), AAI (2), AAII (2); NP-KLH: Ctrl (9), AAI (8), AAII (8)], D NP-specific GC B cells (liveB220+NP+GL7+CD95+) [NP-KLH: Ctrl (10), AAI (12)] by flow cytometry, and (E) GC formation (n = 3) by immunofluorescence staining of spleen section. Scale bar = 50 μm. Each dot represents individual mice and data are pooled from at least 2–3 independent experiments (BD). Data expressed as Mean ± SD. Statistical analyses by One-way ANOVA (B, C) and two-sided t-test (D). C Ctrl vs. AAI ****P < 0.0001, AAI vs. AAII ***P = 0.0007 and Ctrl vs. AAI ****P < 0.0001, AAI vs. AAII **P = 0.0060. D *** P = 0.0008. ns: statistically not significant. Source data are provided as a Source Data file.
Fig. 2
Fig. 2. Mice with kidney disease show defects in affinity maturation, TFh, and secondary B cell response.
C57BL/6 (WT) mice were i.p. injected with a single dose of AAI, AAII (7.5 mg/kg b.wt), or PBS (Ctrl). Mice were either immunized with NP-KLH in alum or left non-immunized (non-immun) 4 days post-AAI injection. At day 12 post-immunization, spleens were assessed for A NP-specific switched IgG1+ B cells (liveIgDIgMCD138Gr1B220+NP+IgG1+) by flow cytometry [Non-immun: Ctrl (3), AAI (3), AAII (3); NP-KLH: Ctrl (9), AAI (8), AAII (7)], B serum NP4 and NP27-specific IgG1 by ELISA [Non-immun: Ctrl (4), AAI (4), AAII (4); NP-KLH Ctrl (12), AAI (8), AAII (10)], and C NP4/NP27-specific IgG1 ratio was determined to measure antibody affinity maturation at day 12 post-immunization [NP-KLH: Ctrl (12), AAI (8), AAII (10)]. D NP7/NP27-specific IgG1 ratio was determined to measure antibody affinity maturation at day 21 post-immunization [NP-KLH: Ctrl (5), AAI (5), AAII (4)]. E NP4-specific antibody-secreting plasma cells generation in the spleen by ELISPOT assay [Non-immun: Ctrl (3), AAI (3), AAII (3); NP-KLH: Ctrl (7), AAI (7), AAII (6)]. F The frequency of TFh (liveCD4+CD44+CXCR5+PD1+) cells in the spleen was determined by flow cytometry at day 12 post-immunization [Non-immun: Ctrl (3), AAI (3), AAII (2); NP-KLH: Ctrl (8), AAI (7), AAII (6)]. AAN and control mice were primed with NP-KLH in alum followed by NP-KLH boost 36 days later. Primed only and boosted mice spleens were evaluated for G total GC (liveB220+GL7+CD95+) [Prime only: Ctrl (7), AAI (7), AAII (7); Prime-boost: Ctrl (10), AAI (14), AAII (8)], H NP-specific switched IgG1+ B cells (liveIgDIgMCD138 Gr1B220+NP+IgG1+) [Prime only: Ctrl (7), AAI (7), AAII (7); Prime-boost: Ctrl (9), AAI (12), AAII (7)], and I NP4-specific IgG1-producing plasma cells 6 days later [Prime only: Ctrl (7), AAI (7), AAII (7); Prime-boost: Ctrl (9), AAI (12), AAII (7)]. Each dot represents individual mice and data are pooled from at least 2–3 independent experiments (AI). Data expressed as Mean ± SD. Statistical analyses by One-way ANOVA (AI). A Ctrl vs. AAI ***P = 0.0007, AAI vs. AAII *P = 0.0252 and Ctrl vs. AAI ****P < 0.0001, AAI vs. AAII ****P < 0.0001. B Ctrl vs. AAI **P = 0.0069, AAI vs. AAII *P = 0.0247. C Ctrl vs. AAI ***P = 0.0004, AAI vs. AAII ***P = 0.0005. D Ctrl vs. AAI *P = 0.0565, AAI vs. AAII **P = 0.0063. E Ctrl vs. AAI *P = 0.020, AAI vs. AAII *P = 0.0299. F Ctrl vs. AAI ***P = 0.0007, AAI vs. AAII *P = 0.0276 and Ctrl vs. AAI **P = 0.0038. G Ctrl vs. AAI ****P < 0.0001, AAI vs. AAII ****P < 0.0001. H Ctrl vs. AAI **P = 0.0038, AAI vs. AAII **P = 0.0073. I Ctrl vs. AAI *P = 0.036, AAI vs. AAII *P = 0.0492. ns: statistically not significant. Source data are provided as a Source Data file.
Fig. 3
Fig. 3. Kidney disease inhibits B cell response during T-dependent immunization.
A Schematic diagram of the experimental plan. AAN and control mice were i.p. immunized with 2.5% SRBC (n = 8) 4 days post-AAI injection. Seven days later, B total GC (liveB220+GL7+CD95+) [SRBC: Ctrl (8), AAI (8), AAII (8)], and C TFh (liveCD4+CD44+CXCR5+PD1+) cells were measured by flow cytometry [SRBC: Ctrl (8), AAI (8), AAII (8)]. AAN mice were either treated with probenecid (PRB + AAI) once on day 0 (relative to AAI injection) or left untreated. Control mice received probenecid only (PRB). Four days later, mice received NP-KLH immunization and assessed for D total GC B cells [PRB (9), AAI (7), PRB + AAI (9)], E TFh cells [PRB (9), AAI (10), PRB + AAI (9)], F NP-specific switched IgG1+ B cells (liveIgDIgMCD138 Gr1B220+ NP+IgG1+) by flow cytometry [PRB (9), AAI (7), PRB + AAI (9)], and G serum NP4-specific IgG1 level by ELISA [PRB (8), AAI (6), PRB + AAI (8)]. H Diagram of the experimental design. WT mice were either subjected to 5/6 nephrectomy (5/6 Nx) (n = 7) or sham-operated (Ctrl) (n = 8). Two weeks after surgery, mice were immunized with NP-KLH and assessed for I total GC [Ctrl (8), 5/6 Nx (7)], J TFh cells [Ctrl (8), 5/6 Nx (7)], K NP-specific switched IgG1+ B cells [Ctrl (8), 5/6 Nx (6)], L serum NP4 and NP27-specific IgG1 levels [Ctrl (8), 5/6 Nx (7)], and M NP4/NP27- IgG1 ratio to measure affinity maturation [Ctrl (8), 5/6 Nx (7)]. Each dot represents individual mice and data are pooled from at least 2–3 independent experiments (BG and IM). Data expressed as Mean ± SD. Statistical analyses by One-way ANOVA (BG) and two-sided t-test (IM). B Ctrl vs. AAI **** P < 0.0001, AAI vs. AAII ****P < 0.0001. C Ctrl vs. AAI **P = 0.0027, AAI vs. AAII *P = 0.0160. D PRB vs. AAI **P = 0.0018, AAI vs. PRB + AAI *P = 0.0472. E PRB vs. AAI *P = 0.0112, AAI vs. PRB + AAI **P = 0.0029. F PRB vs. AAI **P = 0.0012, AAI vs. PRB + AAI *P = 0.0415. G PRB vs. AAI * P = 0.0222, AAI vs. PRB + AAI *P = 0.0484. I **P = 0.0096. J *P = 0.0278. K *P = 0.0295. L **P = 0.0028. M **P = 0.0049. ns: statistically not significant. Source data are provided as a Source Data file.
Fig. 4
Fig. 4. Kidney dysfunction triggers apoptosis in GC B cells.
NP-specific GC B cells from the control and AAN spleen (n = 5) were flow sorted on day 12 post-immunization and subjected to RNA-Seq analysis. A Relative expression of significantly differentially expressed genes (q-value < 0.05) related to cell cycle, and B Over-representation pathway analysis of significantly downregulated genes in AAN GC B cells. C MFI of Ki67+ GC B cells [NP-KLH: Ctrl (5), AAI (7)]. D Cell cycle status of GC B cells was analyzed at day 12 post-immunization [NP-KLH: Ctrl (15), AAI (15)]. E Relative expression of significantly differentially expressed genes (q-value < 0.05) related to apoptosis and F Over-representation pathway analysis of upregulated genes in GC B cells from AAN mice. G NP+ and NP GC B cells staining for active caspase3 expression (n = 12–14) at day 12 post-immunization [NP-KLH: Ctrl (14), AAI (12)]. Relative expression of significantly differentially expressed genes (q-value < 0.05) related to H Light zone (LZ), and I Dark zone (DZ) GC B cells. J Number of LZ centrocytes (CXCR4loCD86hi) and DZ centroblasts (CXCR4hiCD86lo) at day 12 post-immunization [NP-KLH: Ctrl (10), AAI (12)]. Each dot represents individual mice and data pooled from at least 2–3 experiments (C, D, G, and J). Data expressed as Mean ± SD. Statistical analyses by two-sided t-test (C, D, G, and J). C *P = 0.0228. D G0-G1 ***P = 0.0001, S ****P < 0.0001. G **P = 0.0023. J ****P < 0.0001 and ***P = 0.0003. Source data are provided as a Source Data file.
Fig. 5
Fig. 5. HA drives loss of mitochondrial membrane potential in B cells.
A NP-specific GC B cells from the control and AAN spleen (n = 5) were flow sorted on day 12 post-immunization for RNA-Seq analysis. Relative expression of significantly differentially expressed genes (q-value < 0.05) related to mitochondrial membrane potential. At day 12 post-immunization, B mitochondrial membrane potential [NP = KLH: Ctrl (12), AAI (17)], and C mitoROS production [NP = KLH: Ctrl (12), AAI (15)] were measured in GC B cells by TMRE and MitoSox Red staining, respectively. D Schematic diagram of in vitro B cell assay. E WT resting splenic B cells (n = 5) ± uremic toxins were stimulated with αIgM/αCD40/IL-21 and the number of B220+CD138+ plasmablasts was determined by flow cytometry at 96 h. WT B cells ± αIgM/αCD40/IL-21 ± uremic toxins and F proliferation (CFSE dilution) of plasmablasts was assessed at 48 h (n = 3), G apoptosis (n = 6), and H mitochondrial membrane potential was measured in plasmablasts at 24 h (n = 4). I HA concentration in the serum of mice was measured by LC-MS/MS [Ctrl (5), AAI (6)]. J WT splenic B cells + αIgM/αCD40/IL-21 ± HA (35 μg/ml) were assessed for the loss of mitochondrial membrane potential in plasmablasts at 24 h. Each dot represents individual mice (B, C, and I) and 5 (E), 3 (F), 6 (G), and 4 (H, J) experiments. Data pooled from 2–3 experiments (B, C, and I). Data expressed as Mean ± SD. Statistical analyses by One-way ANOVA (EH) and two-sided t-test (B, C, I, and J). B ****P < 0.0001. C **P = 0.0029. E NT vs. GBA ****P < 0.0001, NT vs. IAA **P = 0.0011, NT vs. PC *P = 0.0398, NT vs. PCS *P = 0.0307, NT vs. HA **P = 0.0059, NT vs. Urea **P = 0.0084. G Ctrl vs. HA **P = 0.0019. H Ctrl vs. HA **P = 0.0032. I **P = 0.0037. J ***P = 0.0004. ns: statistically not significant. Source data are provided as a Source Data file.
Fig. 6
Fig. 6. HA causes apoptosis of B cells via GPR109A.
A Resting splenic B cells (n = 5–6) ± αIgM/αCD40/IL-21 or LPS were assessed for Niacr1 mRNA expression by RT-qPCR at 24 h. Total splenocytes from WT and Niacr1/ mice (n = 3) were used as positive and negative controls, respectively. B Niacr1 transcript expression was assessed by RT-qPCR on NP-KLH immunized and FACS-sorted non-GC (liveB220+GL7CD95), total GC (liveB220+GL7+CD95+) (2 mice pooled together) and total spleen cells from WT and Niacr1/ mice (n = 6) at day 12 post-immunization. C Purified total WT B cells from NP-KLH immunized mice (at day 12 post-immunization) were incubated with HA for six hours and assessed for the loss of mitochondrial membrane potential of GC B cells by TMRE staining (B220+GL7+) cells. B cells ± αIgM/αCD40/IL-21± (D and E) GSK256073/± (F) MMF and mitochondrial membrane potential and apoptosis of plasmablasts were measured. Immunized WT mice were i.p. injected with MMF or left untreated (Ctrl) every alternate day and evaluated for G total GC [Non-immun: Ctrl (4), MMF (4); NP-KLH: Ctrl (7), MMF (11)], H NP-specific GC [Non-immun: Ctrl (4), MMF (4); NP-KLH: Ctrl (7), MMF (11)], I serum NP7 and NP27-specific IgG1 titers and affinity maturation (NP7/NP27-IgG1 ratio) [Non-immun: Ctrl (3), MMF (3); NP-KLH: Ctrl (6), MMF (8)] and J loss of mitochondrial membrane potential of GC B cells [Non-immun: Ctrl (4), MMF (5); NP-KLH: Ctrl (8), MMF (13)]. K Resting splenic B cells (n = 3) from WT and Niacr1/ mice were stimulated with αIgM/αCD40/IL-21 in the presence or absence of HA. The B220+CD138+ cells were analyzed for the loss of mitochondrial membrane potential by TMRE staining 24 h later. Each dot represents individual mice (B and GJ) and individual experiments (A, CF, and K). Data pooled from 2–3 independent experiments (B and GJ) and individual experiments A (5), C, D (8), E, F (7) and K (3). Data expressed as Mean ± SD. Statistical analyses by One-way ANOVA (A, B, and DK) and two-sided t-test (C). A Resting vs. WT spleen **P = 0.0015, αIgM/αCD40/IL-21 vs. WT spleen **P = 0.0010, LPS vs WT spleen *P = 0.0523, WT spleen vs. Niacr1/ spleen ****P < 0.0001. B Non-GC B cells ***P = 0.0005, GC B cells ***P = 0.0004, Total Spleen ***P = 0.0002. C ***P = 0.0004 and ****P = 0.0005. D ****P < 0.0001. E ****P < 0.0001. F ***P = 0.0008. G ****P < 0.0001. H ***P = 0.0002. I **P = 0.0067 and NP7/NP27: *P = 0.0106. J **P = 0.0015. K **P = 0.0134. ns: statistically not significant. Source data are provided as a Source Data file.
Fig. 7
Fig. 7. B cell-specific GPR109A expression induces apoptosis in kidney disease.
A Diagram of the experimental design. WT and Niacr1/ mice (n = 8–10) were subjected to AAN followed by NP-KLH in alum immunization. B Total GC B cells (liveB220+GL7+CD95+) [NP-KLH: WT Ctrl (10), AAI (8); Niacr1/ Ctrl (9), AAI (10)], C NP-specific GC B cells (liveB220+NP+GL7+CD95+) by flow cytometry [NP-KLH: WT Ctrl (11), AAI (9); Niacr1/ Ctrl (11), AAI (10)], and D antibody affinity maturation as assessed by NP7/NP27-specific serum IgG1 ratio was evaluated at day 12 post-immunization [NP-KLH: WT Ctrl (6), AAI (4); Niacr1/ Ctrl (5), AAI (6)]. E Resting splenic B cells from WT or Niacr1/ mice were adoptively transferred into control or AAN μMT mice one day before AAI injection. Mice were immunized with NP-KLH in alum four days post-AAI-injection. At day 12 post-immunization, spleens were assessed for F total GC formation [NP-KLH: WT Ctrl (8), AAI (12); Niacr1/ Ctrl (10), AAI (10)], G NP-specific GC formation [NP-KLH: WT Ctrl (4), AAI (5); Niacr1/ Ctrl (5), AAI (6)], H proliferation [NP-KLH: WT Ctrl (4), AAI (5); Niacr1/ Ctrl (5), AAI (6)], I loss of mitochondrial membrane potential [NP-KLH: WT Ctrl (4), AAI (5); Niacr1/ Ctrl (5), AAI (6)], J mitochondrial ROS generation by total GC B cells [NP-KLH: WT Ctrl (4), AAI (5); Niacr1/ Ctrl (5), AAI (6)], K apoptotic NP-specific GC B cells by flow cytometry [NP-KLH: WT Ctrl (4), AAI (5); Niacr1/ Ctrl (5), AAI (6)], and L antibody affinity maturation as assessed by NP7/NP27-specific serum IgG1 ratio [NP-KLH: WT Ctrl (4), AAI (6); Niacr1/ Ctrl (4), AAI (5)]. Each dot represents individual mice (BD and FL). Data pooled from 2–3 independent experiments (BD, and FL). Data expressed as Mean ± SD. Statistical analyses by One-way ANOVA (BD, and FL). B WT Ctrl vs AAI ****P < 0.0001, WT AAI vs Niacr1/ AAI **P = 0.0017. C WT Ctrl vs AAI **P = 0.005, WT AAI vs Niacr1/ AAI *P = 0.04852. D WT Ctrl vs AAI **P = 0.0060. F WT Ctrl vs AAI ****P < 0.0001, WT AAI vs Niacr1/ AAI *P = 0.0142. G WT Ctrl vs AAI ****P < 0.0001, WT AAI vs Niacr1/ AAI **P = 0.006. H WT Ctrl vs AAI **P = 0.0093, WT AAI vs Niacr1/ AAI **P = 0.0018. I WT Ctrl vs AAI *P = 0.0162, WT AAI vs Niacr1/ AAI **P = 0.0010. J WT Ctrl vs AAI *P = 0.0243, WT AAI vs Niacr1/ AAI *P = 0.0110. K WT Ctrl vs AAI **P = 0.0096, WT AAI vs Niacr1/ AAI *P = 0.0115. L WT Ctrl vs AAI *P = 0.0653, WT AAI vs Niacr1/ AAI *P = 0.0150. ns: statistically not significant. Source data are provided as a Source Data file.
Fig. 8
Fig. 8. Diminished B and T cells response in influenza-infected mice in kidney disease.
A Schematic representation of the experimental design. Mice were either injected with AAI or PBS (Ctrl). Four days post-AAI injection, mice were infected with 1000 PFU of mouse-adapted influenza H1N1 strain A/PR/8/34 via oropharyngeal aspiration (n = 7–9) or left sham infected (n = 3). At day 12 p.i., spleen of infected and sham mice was assessed for B GC (liveB220+GL7+CD95+) [Sham: Ctrl (3), AAI (2); Influenza: Ctrl (7), AAI (9)], and C TFh (liveCD4+CD44+CXCR5+PD1+) cells by flow cytometry [Sham: Ctrl (3), AAI (2); Influenza: Ctrl (7), AAI (9)], D serum anti-PR8HA IgG titer by HAI assay [Sham: Ctrl (1), AAI (1); Influenza: Ctrl (8), AAI (7)], and E influenza-specific CD8+ T cells by tetramer staining [Sham: Ctrl (2), AAI (2); Influenza: Ctrl (7), AAI (7)]. F Influenza A virus infected and sham lungs were subjected to H&E staining and peri-vascular, peri-bronchiolar, and parenchymal inflammation were scored [Sham: Ctrl (1), AAI (1); Influenza: Ctrl (7), AAI (8)]. Scale bar = 50 μm. G Viral load in the lung was quantified by assessing Influenza A virus M1 gene expression by Taqman qPCR assay [Influenza: Ctrl (5), AAI (9)]. Data is normalized to the HPRT gene. Each dot represents individual mice and data are pooled from 2–3 independent experiments (BG). Data expressed as Mean ± SD. Statistical analyses by One-way ANOVA (B, C, and E), two-sided t-test (F), Mann–Whitney test (G), and Kruskal–Wallis test (D). B ***P = 0.0007. C ***P = 0.0003. D ***P = 0.0002. E ***P = 0.0005. F Peri-vascular *P = 0.020521. G **P = 0.007. Source data are provided as a Source Data file.

References

    1. Valderas, J. M., Starfield, B., Sibbald, B., Salisbury, C. & Roland, M. Defining comorbidity: implications for understanding health and health services. Ann. Fam. Med.7, 357–363 (2009). - PMC - PubMed
    1. Gijsen, R. et al. Causes and consequences of comorbidity: a review. J. Clin. Epidemiol.54, 661–674 (2001). - PubMed
    1. Kovesdy, C. P. Epidemiology of chronic kidney disease: an update 2022. Kidney Int. Suppl. (2011)12, 7–11 (2022). - PMC - PubMed
    1. Mahalingasivam, V. et al. COVID-19 and kidney disease: insights from epidemiology to inform clinical practice. Nat. Rev. Nephrol.18, 485–498 (2022). - PMC - PubMed
    1. Brogan, M. & Ross, M. J. COVID-19 and kidney disease. Annu Rev. Med74, 1–13 (2023). - PubMed

Publication types

Associated data