Andrew J. Kassianos’s research while affiliated with Royal Brisbane Hospital and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (82)


Automated 3D Segmentation of Glomeruli in Human Kidney Tissue Specimens Using 16.4 T MRI Without Contrast Agents
  • Conference Paper

April 2025

·

3 Reads

Aurel J. Amar

·

·

·

[...]

·

David C. Reutens

Head-to-head comparison of tyrosine kinase inhibitors in renal cell carcinoma using patient-derived cell culture

March 2025

·

3 Reads

Background: Metastatic renal cell carcinoma (RCC) is often treated with a combination of immunotherapy and tyrosine kinase inhibitors (TKIs). Patient-derived RCC cells were cultured and inter-individual differences to treatment with a panel of TKIs were evaluated. Methods: Tumor tissue was collected during nephrectomy. Cells were cultured and treated with a panel of clinically relevant TKIs (sunitinib, cabozantinib, pazopanib, axitinib) at concentrations of 5 µM for 48-72 hours. Cell viability was evaluated using MTT assays. One-sided T-tests were used to evaluate results. Results: Patient-derived cancer cells were able to be grown beyond 10 passages from 12/38 samples collected (27%). Four patient-derived samples were tested against the TKI panel. No substantial difference between drugs was seen for two samples. In one sample, there was a clear superior response to sunitinib (48% mean viability, vs >75% for the other drugs). For the final sample, sunitinib, cabozantinib, and axitinib demonstrated a superior response compared with pazopanib (71%, 77%, 70%, and 85% mean viability, respectively). Conclusions: Inter-individual variability in the responses of patient-derived RCC cultures to TKIs was seen, which may have biological and clinical significance. Future directions could build on this work to develop personalized cancer susceptibility profiles, with potential for translation into a clinical trial.


Urinary EGF: Back to the Future
  • Article
  • Full-text available

March 2025

·

1 Read

Kidney360

Download

TGF‐β1 increased expression of fibrogenesis‐associated genes. Using qPCR, relative expression of target genes to the mean of the reference gene (18 s rRNA) was measured. Target genes investigated were (a, b) extracellular matrix proteins fibronectin (FN) and collagen type IV alpha 1 (COL4A1); (c, d) enzymes involved in fatty acid oxidation pathway, peroxisomal acyl‐coenzyme A oxidase 1 (ACOX1) and acetyl‐CoA acyltransferase 1 (ACAA1); and (e) mesenchymal cell‐associated protein alpha smooth muscle actin (ACTA2). They were compared at 24, 48, and 72 h at 1, 5, and 10 ng/mL TGF‐β1. Shaded columns and timepoint indicate the TGF‐β1 concentration and treatment time selected for downstream NMR analysis. Gene expression is shown as a mean fold change ± SD from vehicle control (0 ng/mL). Individual significant comparisons (compared to the 0 ng/mL) are annotated. n = 4 for FN and ACTA2, n = 3 for ACOX1 and COL4A1, n = 2 for ACAA1.
TGF‐β1 increased fibronectin and PAI‐1 protein in culture supernatant. Cell supernatant was obtained from cultured HK‐2 cells at different timepoints following treatment (24, 48, and 72 h) with TGF‐β1 (vehicle control 0; 1, 5, and 10 ng/mL). Representative Western blots are demonstrated in (a), the protein ladder is represented along the left‐hand side. The protein band intensity for PAI‐1 (b) and fibronectin (c) was normalized to total protein loading (Ponceau S) for each sample, and within each timepoint, all samples were normalized to the vehicle control. Data were logarithmically transformed and analysed using two‐way ANOVA with Tukey's multiple comparisons test. Bars show mean fold change in band intensity ± SD (n = 4). *p < 0.05; **p < 0.01.
Representative 2D COSY spectra from vehicle control and TGF‐β1‐treated HK‐2 cells. Examples show two‐dimensional correlated spectroscopy (2D‐COSY) for (a) vehicle control (0 ng/mL) and (b) TGF‐β1 treatment (10 ng/mL) at 48 h treatment.
Plots of spectral data using PCA and OPLS‐DA. (a) Clustering by principal components analysis (PCA) is demonstrated for vehicle control (0 ng/mL; green) and TGF‐β1 treatment (10 ng/mL; blue). In (b) orthogonal projections to latent structures – discriminant analysis (OPLS‐DA) plots demonstrate clear separation of control and treated groups using supervised assignment of components.
Metabolite pathway alterations identified by magnetic resonance metabolomics in a proximal tubular epithelial cell line treated with TGF‐β1

February 2025

·

6 Reads

Tubulointerstitial fibrosis is a characteristic hallmark of chronic kidney disease (CKD). Metabolic perturbations in cellular energy metabolism contribute to the pathogenesis of CKD, but the chemical contributors remain unclear. The aim of this investigation was to use two dimensional ¹H‐nuclear magnetic resonance (2D‐COSY) metabolomics to identify the chemical changes of kidney fibrogenesis. An in vitro transforming growth factor‐β1 (TGF‐β1)‐induced model of kidney fibrogenesis with human kidney‐2 (HK‐2) proximal tubular epithelial cells (PTEC) was used. The model was validated by assaying for various pro‐fibrotic molecules, using quantitative PCR and Western blotting. 2D‐COSY was performed on treated cells. Morphological and functional changes characteristic of tubulointerstitial fibrosis were confirmed in the model; expression of fibronectin, collagen type IV, smooth muscle actin, oxidative stress enzymes increased (p < 0.05). NMR metabolomics provided evidence of altered metabolite signatures associated with glycolysis and glutamine metabolism, with decreased myo‐inositol and choline, and metabolites of the oxidative phase of the pentose phosphate pathway with increased glucose and glucuronic acid. The altered PTEC cellular metabolism likely supports the rapid fibrogenic energy demands. These results, using 2D‐COSY metabolomics, support development of a biomarker panel of fibrosis detectable using clinical magnetic resonance spectroscopy to diagnose and manage CKD.


Paediatric screening recommendations from major cardiovascular and lipid societies.
Paediatric screening strategies for FH.
Universal Paediatric and Newborn Screening for Familial Hypercholesterolaemia—Challenges and Opportunities: An Australian Perspective

February 2025

·

58 Reads

Lipidology

Heterozygous familial hypercholesterolaemia is one of the most common genetic conditions leading to premature atherosclerotic cardiovascular disease. It can be diagnosed using a combination of clinical, biochemical, and genetic tools. Most guidelines recommend screening during childhood and treatment from the age of 8–10 years. However, screening remains sporadic in most countries and the majority of individuals remain undiagnosed. Registry studies have highlighted the ongoing delayed and low percentage of detection of FH in children. Universal early childhood screening models utilising a combination of biomarker-based and genetic testing have been trialled and are in practice in some countries. Newborn screening is a public health success story and one of the most effective public health measures. It offers universal screening for conditions that can result in significant morbidity or even death if left untreated. There has been renewed interest in including familial hypercholesterolaemia in newborn screening programmes. Using cord blood to identify familial hypercholesterolaemia has not yielded convincing results. However, novel screening approaches on dried blood spots that include biomarker-based lipid profile testing alone, in combination with confirmatory genetic testing, or first-line genetic testing have shown promising results. This provides the opportunity of early diagnosis and treatment of infants and their extended families. However, challenges are associated with the inclusion of familial hypercholesterolaemia in newborn screening programmes with significant impacts on the newborn, family members, and public health.


Fig. 2 Hypoxia and IL-1β synergistically down-regulate cell cycling pathways in human primary PTECs. A Significantly (FDR < 0.05) enriched KEGG pathways identified from 'unique' down-regulated differentially expressed genes (DEGs) for the Hypoxia+IL-1β vs. Normoxia+Vehicle contrast. The count (represented by point size) is the number of DEGs annotated in the enriched KEGG pathway. The gene ratio (x-axis) is the count divided by the total number of DEGs with annotations across the collection of KEGG pathways. FDR is represented by point colour. B Heatmap showing normalised gene expression values (scaled and centred) for unique DEGs from the Hypoxia+IL-1β vs. Normoxia+Vehicle contrast contributing to the KEGG Cell cycle pathway. Expression values for all samples are shown, with samples (columns) ordered by treatment then patient. C Scatterplot of normalised expression values (log 2 CPM) for CCNA2, CCNB1 and CCNB2 across all PTEC samples. D Top panel: Fold changes (relative to Normoxia+Vehicle) in Cyclin A2 protein levels (as a ratio of loading control β-actin) for PTECs cultured for 48 h under normoxic (norm) or hypoxic (hypox) conditions in the absence (vehicle; veh) or presence of IL-1β. Bar graphs represent mean ± SEM. Symbols represent individual donor PTECs; n = 4. *P < 0.05, **P < 0.01, one-way ANOVA with Tukey's multiple-comparison test. Bottom panel: Cyclin A2 Western blot for PTECs cultured under normoxic (norm) or hypoxic (hypox) conditions in the absence (vehicle; veh) or presence of IL-1β (20 µg total protein per lane). Representative images from one of four donor PTECs are presented. Full and uncropped Western blot available as Supplementary Material.
Fig. 4 Significantly elevated PTEC IL-1RI expression and senescence in fibrotic kidneys. A Left panel: Quantitative analysis of % IL-1RI + PTECs (proportion of AQP-1 + cells (i.e., PTECs) expressing IL-1RI) in control (non-fibrotic; n = 4) and fibrotic kidney tissue (n = 5). Points represent values for individual donor tissue samples. Results represent mean ± SEM of values from four randomly selected areas for each tissue sample. *P < 0.05, Welch's t-test. Right panel: Representative immunofluorescent labelling of control (non-fibrotic) and fibrotic kidney tissue stained for PTEC marker aquaporin-1 (AQP-1; white), IL-1RI (green) and DAPI (blue). Scale bars represent 20 µm. IL-1RI + PTECs are highlighted with white arrows. B Left panel: Quantitative analysis of % SA-β-gal + PTECs (proportion of AQP-1 + cells (i . e., PTEC) with positive Spider-β-gal staining) in control (non-fibrotic; n = 4) and fibrotic kidney tissue (n = 4). Points represent values for individual donor tissue samples. Results represent mean ± SEM of values from four randomly selected areas for each tissue sample. **P < 0.01, Welch's t-test. Right panel: Representative immunofluorescent images of control (non-fibrotic) and fibrotic kidney tissue labelled with PTEC marker AQP-1 (white), Spider-β-gal (green) and DAPI (blue). Scale bars represent 20 µm. SA-β-gal + PTECs are highlighted with white arrows. Lower magnification immunofluorescent images are presented in Fig. S4.
Human proximal tubular epithelial cell interleukin-1 receptor signalling triggers G2/M arrest and cellular senescence during hypoxic kidney injury

January 2025

·

33 Reads

·

1 Citation

Cell Death and Disease

Hypoxia and interleukin (IL)-1β are independent mediators of tubulointerstitial fibrosis, the histological hallmark of chronic kidney disease (CKD). Here, we examine how hypoxia and IL-1β act in synergy to augment maladaptive proximal tubular epithelial cell (PTEC) repair in human CKD. Ex vivo patient-derived PTECs were cultured under normoxic (21% O 2 ) or hypoxic (1% O 2 ) conditions in the absence or presence of IL-1β and examined for maladaptive repair signatures. Hypoxic PTECs incubated with IL-1β displayed a discrete transcriptomic profile distinct from PTECs cultured under hypoxia alone, IL-1β alone or under normoxia. Hypoxia+IL-1β-treated PTECs had 692 ‘unique’ differentially expressed genes (DEGs) compared to normoxic PTECs, with ‘cell cycle’ the most significantly enriched KEGG pathway based on ‘unique’ down-regulated DEGs (including CCNA2 , CCNB1 and CCNB2 ). Hypoxia+IL-1β-treated PTECs displayed signatures of cellular senescence, with reduced proliferation, G2/M cell cycle arrest, increased p21 expression, elevated senescence-associated β-galactosidase (SA-β-gal) activity and increased production of pro-inflammatory/fibrotic senescence-associated secretory phenotype (SASP) factors compared to normoxic conditions. Treatment of Hypoxia+IL-1β-treated PTECs with either a type I IL-1 receptor (IL-1RI) neutralizing antibody or a senolytic drug combination, quercetin+dasatinib, attenuated senescent cell burden. In vitro findings were validated in human CKD bio-specimens (kidney tissue, urine), with elevated PTEC IL-1RI expression and senescence (SA-β-gal activity) detected in fibrotic kidneys and numbers of senescent (SA-β-gal ⁺ ) urinary PTECs correlating with urinary IL-1β levels and severity of interstitial fibrosis. Our data identify a mechanism whereby hypoxia in combination with IL-1β/IL-1RI signalling trigger PTEC senescence, providing novel therapeutic and diagnostic check-points for restoring tubular regeneration in human CKD.


Nephron Senescence and Mechanisms

December 2024

·

18 Reads

Kidney aging is the loss of tissue and organ function over time. Cellular senescence is a state of permanent growth arrest and cessation of cell division. Although aging and senescence share some features, the two processes differ. In contrast to aging, senescence occurs when a cell irreversibly exits the cell cycle and assumes a distinct phenotype. This chapter on nephron senescence and mechanisms explores the proposition that senescence in the kidney is more than just normal organ aging but instead encompasses pathobiological pathways that are activated by injury and result in pathologic alterations in kidney tissue and function. We discuss the role of cellular senescence in a variety of kidney diseases and consider the potential therapeutic benefits of targeting senescent cells in diseased and aged kidneys.


Fabry Disease Podocytes Reveal Ferroptosis as a Potential Regulator of Cell Pathology

November 2024

·

18 Reads

·

3 Citations

Kidney International Reports

Introduction Fabry disease (FD) results from pathogenic GLA variants, leading to a deficiency in lysosomal α-galactosidase A (α-Gal A) and accumulation of the sphingolipid globotriaosylceramide (Gb3). This leads to severe renal and cardiovascular complications, primarily affecting kidney podocytes. As a multisystemic disorder, FD initiates at the cellular level; however, the mechanism(s) underlying Gb3-induced cell dysfunction remain largely unknown. This study aimed to identify potential drivers of FD and explore the underlying cell pathology in induced pluripotent stem cell (iPSC)–derived podocytes from patients with FD. Methods iPSCs were derived from patients with FD with GLAc.851T>C or GLAc.1193_1196del variants and compared with controls or CRISPR-Cas9–corrected cell lines. iPSCs were differentiated into podocytes; and α-Gal A activity, Gb3 accumulation, and cell morphology were assessed. Label-free mass spectrometry identified the top, differentially expressed proteins which were validated by using western blot. Results Podocytes derived from patients with FD exhibited expression of podocyte-specific markers and morphological features of FD. Reduced α-Gal A activity was observed in FD iPSC-derived podocytes along with the accumulation of Gb3. Proteomic profiling revealed distinct proteomic signatures between control and iPSC-derived podocytes from a patient with FD, with apparent variations among FD lines, highlighting GLA variant–specific proteomic alterations. Notably, the ferroptosis-associated protein, arachidonate 15-lipoxygenase (ALOX15), was the most upregulated protein in FD podocytes and ferroptosis was the most enriched pathway. Western blot analysis confirmed the upregulation of ALOX15 in FD podocytes, with validation of other markers implicating ferroptosis in FD pathology. Conclusion These findings underscore the heterogeneity of FD and, for the first time, implicate ferroptosis as a potential common pathway driving its pathology.


Signalling cascades of discrete modes of RCD. Apoptosis: (1a) The extrinsic apoptosis pathway is initiated following activation of death ligands [e.g., tumour necrosis factor receptor (TNFR) or TNF-related apoptosis-inducing ligand receptors (TRAILR)] by their respective ligands, which in turn triggers (1b) proteolytic cleavage of cysteine proteases, named caspases, culminating in the maturation of caspase-7 (CASP7) and caspase-3 (CASP3). (1c) Active caspase-3 proteolytically cleaves a range of targets, including (1d) ROCK1 which triggers membrane “blebbing” (1e) and apoptotic cell death. (1f) Additionally, active caspase-3 mediates the cleavage of “flippases”, thus permitting phosphatidylserine presentation on the cell membrane outer leaf. (1g) Active caspase-3 also interferes with anti-apoptotic signalling through cleavage of the p65 subunit of nuclear factor-κB (NF-κB). Mitochondrial permeability transition pore (mPTP)-necrosis: (2a) An accumulation of mitochondrial reactive oxygen species (ROS; e.g., ↑O2 ·) and calcium (Ca²⁺) trigger the opening of the mPTP. (2b) The F1F0 subunit of ATP synthase forms part of the inner mitochondrial membrane pore as a component of a “synthasome” complex, consisting of PPIF/cyclophilin D (CypD), ANT and PiC. (2c) The BAX and BAK proteins which accumulate in regions surrounding the mPTP are hypothesised to form part of the outer pore on the cytosolic leaf of the mitochondrial membrane. (2d) Once formed, the mPTP facilitates the release of protons (H⁺) and cytochrome c (CytoC) from the mitochondrial matrix into the cell. Proton release depolarizes the mitochondrial membrane (↓∆Ψ mt ), preventing further ATP and NAD⁺ biosynthesis. (2e) The combination of these leads to failed homeostasis and necrotic cell death. Necroptosis: (3a) Activation of TNFR, TRAILR and/or type I interferon receptors (IFNR) mediate the phosphorylation of RIPK-1 and -3, which in turn accumulate into structures called “necrosomes” (3b). (3c-d) Alternative pathways of RIPK3 activation are also identified, including binding to TRIF in response to pattern recognition receptor (PRR) engagement and Z-DNA-binding protein 1 (ZBP1) activated by viral or cellular RNA. (3e) Phosphorylated-RIPK3, in turn, mediates the phosphorylation of MLKL (pMLKL). (3f) pMLKL accumulates on the inner leaf of the cell membrane near Zonula Occludens-1 (ZO-1). (3g) These “hotspots” of pMLKL are hypothesised to overcome a physical membranolytic threshold, which causes membrane blow-out and the lytic release of the cytosol to the extracellular space. (3h) Phosphorylated RIPK3 also interacts with NF-κB signalling components to drive pro-inflammatory cytokine release. Ferroptosis: (4a) The accumulation of free iron (Fe²⁺) and ROS (e.g., from the mitochondria) drives the oxidation of phospholipids and the formation of phospholipid peroxyl radicals (PLOO·) (4b). These lipid peroxyl radicals propagate the oxidation of additional phospholipids, which, if not terminated or repaired, will ultimately cause the loss of cell membrane integrity (4c). Repair of oxidised phospholipids may occur via two pathways: the first (4d) involves ferroptosis suppressor protein 1 (FSP1) converting phospholipid peroxyl radicals (PLOO·) to phospholipid hydroperoxides (PLOOH). (4e) The second pathway of lipid repair involves glutathione peroxidase 4 (GPX4) reducing in a glutathione (GSH)-dependent reaction phospholipid hydroperoxides (PLOOH) to phospholipid alcohols (PLOH) (4f). (4g) System Xc⁻ consists of a heavy chain component, 4F2hc (SLC3A2) and a transport module, xCT (SLC7A11), and functions as a dedicated cysteine import system, which is an important precursor of glutathione.
NLRP3 inflammasome priming and activation. (A) Signal 1: Priming - Activation of membrane-bound and cytosolic pattern recognition receptors [toll-like receptors (TLRs) by DAMPs/PAMPs (e.g., lipopolysaccharide; LPS), interleukin-1 receptors (IL-1R) by IL-1β, or tumour necrosis factor (TNF) receptors by TNF cytokines] triggers a nuclear factor-κB (NF-κB) signalling cascade, which promotes the transcription of inflammasome components, including NLRP3 and the pro-inflammatory IL-1 family cytokines, IL-1β and IL-18. (B) Signal 2: Activation - PAMP and/or DAMP [e.g., extracellular ATP, pore-forming toxins (nigericin), or particular matter] trigger intracellular pathways (mitochondrial dysfunction, oxidative stress) that “activate” the formation of the NLRP3 inflammasome “disk”, a multimeric oligomeric signalling platform, facilitated by the accessory NIMA-related kinase 7 (NEK7) protein. The adaptor protein ASC [apoptosis-like speck-domain containing a caspase activation and recruitment domain (CARD)] is recruited to the inflammasome, and, in-turn, interacts with caspase-1 through homodimeric interactions. Recruitment of caspase-1 to the inflammasome complex mediates the dimerization, autolytic cleavage and activation of caspase-1. Once activated, caspase-1 cleaves pore-forming protein, gasdermin D (GSDMD), and the proinflammatory cytokines, IL-1β and IL-18. Cleaved-GSDMD and active IL-1β/IL-18 accumulate at the cell membrane, where GSDMD forms pores in the cell membrane that facilitate cell lysis/pyroptosis and IL-1β/IL-18 release. (C) Caspase-8 activation can also cause pyroptosis either directly through its recruitment into the inflammasome complex or via its cleavage of GSDMD or indirectly by activation of caspase-3 to cleave gasdermin E (GSDME).
Regulated cell death in chronic kidney disease: current evidence and future clinical perspectives

October 2024

·

15 Reads

·

1 Citation

Chronic kidney disease (CKD) is the progressive loss of kidney function/structure over a period of at least 3 months. It is characterised histologically by the triad of cell loss, inflammation and fibrosis. This literature review focuses on the forms of cell death that trigger downstream inflammation and fibrosis, collectively called regulated cell death (RCD) pathways. Discrete forms of RCD have emerged as central mediators of CKD pathology. In particular, pathways of regulated necrosis – including mitochondrial permeability transition pore (mPTP)-mediated necrosis, necroptosis, ferroptosis and pyroptosis – have been shown to mediate kidney pathology directly or through the release of danger signals that trigger a pro-inflammatory response, further amplifying tissue injury in a cellular process called necroinflammation. Despite accumulating evidence in pre-clinical models, no clinical studies have yet targeted these RCD modes in human CKD. The review summarizes recent advances in our understanding of RCD pathways in CKD, looks at inter-relations between the pathways (with the emphasis on propagation of death signals) and the evidence for therapeutic targeting of molecules in the RCD pathways to prevent or treat CKD.



Citations (44)


... Integrating data from comprehensive diagnostic tools, including genomics, transcriptomics, proteomics, and metabolomics, can facilitate diagnosis. Proteomics data specifically can also lead to better understanding of disease mechanism and potential therapeutic targets, exemplified by recent work in Fabry disease [2,3]. Unfortunately, many of these tools including proteomics remain available only in the research setting, significantly limiting the reach to many people with rare disease and their families [4]. ...

Reference:

The case for including proteomics in routine diagnostic practice for rare disease
Fabry Disease Podocytes Reveal Ferroptosis as a Potential Regulator of Cell Pathology
  • Citing Article
  • November 2024

Kidney International Reports

... DAMPs such as NLRP3 inflammasome and Toll-like receptors are elicited by renal resident cells, specifically tubular epithelial cells and podocytes, of which are known to be injured. These pathways include NF-κB, JAK-STAT, and cGAS-STING which in turn induces overexpression of pro-inflammatory cytokines (TNFα, IL-1β), and even additional chemokines that attract monocytes and T lymphocytes into the interstitium (Yuan et al., 2022;Giuliani et al., 2024). This increased inflammation drives further immunological instability which further aggravates the tumor microenvironment through incessant signals perpetuating inflammation thereby increasing M1 macrophage activity. ...

Regulated cell death in chronic kidney disease: current evidence and future clinical perspectives

... This leads to an increase in BACH2 O-GlcNAcylation in neighboring cells, transcriptionally suppressing proteins that protect against ferroptosis such as SCL7A11 and GPX4 [28]. This propagation through IRI-sEVs also may play a role in the transition of AKI-to-CKD [29]. ...

Human proximal tubular epithelial cell-derived small extracellular vesicles mediate synchronized tubular ferroptosis in hypoxic kidney injury
  • Citing Article
  • January 2024

Redox Biology

... SPP1 + TAMs colocalized with fibroblast activation protein (FAP) + fibroblasts, and high expression of SPP1 or FAP correlated with diminished benefits from immunotherapy (104). Studies on clear cell renal cell cancer suggested that exhausted immune cells and the lack of PD-1, PD-L1, and CTLA-4 expressions could underline the non-responsiveness to immunotherapy (105). ...

High risk clear cell renal cell carcinoma microenvironments contain protumour immunophenotypes lacking specific immune checkpoints

npj Precision Oncology

... While surgical resection remains primary treatment for localized cases, ~ 30% experience recurrence and high mortality upon metastasis [5,6]. Despite recent survival improvements through targeted therapies and immunotherapy, challenges persist in understanding tumor heterogeneity, immune evasion mechanisms, and personalizing treatment strategies [7,8]. This underscores the critical need to identify novel biomarkers and therapeutic targets for ccRCC. ...

Cellular milieu in clear cell renal cell carcinoma

... Ferroptosis is not only a key factor leading to abnormal inflammatory responses but also indirectly participates in renal fibrosis by promoting the infiltration of inflammatory cells and the release of inflammatory mediators [102][103][104]. Studies have shown that ferroptosis can activate the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, promoting the release of pro-inflammatory factors such as interleukin-1β (iL-1β) and interleukin-18 (iL-18), exacerbating renal tissue inflammation, and thereby aggravating renal fibrosis [105,106]. Ferroptosis promotes the polarization of macrophages toward the m1 phenotype, and m1 macrophages secrete large amounts of pro-inflammatory factors, further promoting fibrosis [104,107]. ...

Hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c+ dendritic cells

Cell Death and Disease

... The limitations of scRNA-seq highlight the urgent need for advanced spatially resolved transcriptomics approaches that integrate gene expression data with the spatial context of renal cellular architecture. Techniques such as Slide-seqV2 and other high-resolution spatial transcriptomics methods have demonstrated the potential to uncover disease-specific cell neighborhoods and pathways, thereby providing a more comprehensive understanding of kidney pathology [111,112]. These methodologies allow for the mapping of cellular interactions and the identification of transcriptional signatures specific to various nephron segments, which is essential for elucidating the complex interplay of cells within the kidney [113]. ...

Spatially Resolved Transcriptomes of Mammalian Kidneys Illustrate the Molecular Complexity and Interactions of Functional Nephron Segments

... Preclinical and in-vitro reports on plant-based therapies demonstrate robust therapeutic benefits of plant-derived molecules against CKD [19]. Natural plant compounds are active ingredients that have been extracted from plants and can modulate several pathways, including anti-inflammatory, anti-fibrotic, and anti-oxidant actions [20]. ...

Promoting Plant-Based Therapies for Chronic Kidney Disease

Journal of Evidence-Based Integrative Medicine

... Orally ingested adenine tends to accumulate more extensively than other purines and has been frequently used to induce gradual kidney damage [38]. Adenine is rapidly metabolized to DHA, leading to crystal formation in the proximal tubule, which closely resembles human CKD [39]. Adenine treatment resulted in increased levels of kidney function biomarkers and pro brogenic markers [40,41]. ...

Adenine overload induces ferroptosis in human primary proximal tubular epithelial cells

Cell Death and Disease

... While rare cases of NS secondary to minimal change disease or CG have been reported after the recent COVID-19 vaccination, a causal relationship remains unclear. 38,39 HISTOLOGICAL FEATURES OF HIVAN AND COVAN HIV and SARS-CoV-2 cause a diverse spectrum of kidney pathology beyond CG. For an excellent review of glomerular diseases in persons with HIV, refer to the 2018 Kidney Disease Improving Global Outcomes Global Controversies Conference on "Kidney disease in the setting of HIV infection". ...

SARS-CoV-2 vaccination–associated collapsing glomerulopathy in a kidney transplant recipient
  • Citing Article
  • January 2022

Kidney International