Indian Journal of Animal Research

  • Chief EditorM. R. Saseendranath

  • Print ISSN 0367-6722

  • Online ISSN 0976-0555

  • NAAS Rating 6.40

  • SJR 0.233, CiteScore: 0.606

  • Impact Factor 0.4 (2024)

Frequency :
Monthly (January, February, March, April, May, June, July, August, September, October, November and December)
Indexing Services :
Science Citation Index Expanded, BIOSIS Preview, ISI Citation Index, Biological Abstracts, Scopus, AGRICOLA, Google Scholar, CrossRef, CAB Abstracting Journals, Chemical Abstracts, Indian Science Abstracts, EBSCO Indexing Services, Index Copernicus

Galngin Mitigates Glycerol-induced Rhabdomyolysis-associated Acute Kidney Injury by Modulating Nf-κb-mediated Cytokine Pathways and Apoptotic Responses

Manal F. El-Khadragy1, Rewaida Abdel-Gaber2, Nourhan H. Mohamed3, Fatma Elzahraa H. Salem4,*
  • https://orcid.org/0000-0001-5457-6791, https://orcid.org/0000-0001-9263-6871, https://orcid.org/0009-0005-7002-6366, https://orcid.org/0000-0002-4334-2081
1Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
2Department of Zoology, College of Science, King Saud University, P.O. 2455, Riyadh 11451, Saudi Arabia.
3Faculty of Medicine, Newgiza University, New Giza, Cairo, Egypt.
4Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt, P.O. Box 11611, Egypt.

Background: This study investigated the protective capabilities of galangin, a naturally occurring flavonoid found in galangal and propolis, against glycerol-induced acute kidney injury (AKI), a serious complication associated with rhabdomyolysis.

Methods: Rats were pre-treated with galangin (100 mg/kg) for 21 days before receiving a glycerol single injection (50%, 10 mg/kg, intramuscular) to induce AKI. According to the findings, the kidneys in the AKI group showed notable molecular, and functional alterations.

Result: Treatment with galangin notably reduced kidney relative weight and decreased serum levels of urea and creatinine, indicating preservation of renal function. Furthermore, the intervention lowered concentrations of lactate dehydrogenase (LDH) and creatine kinase (CK), key indicators of muscle breakdown associated with rhabdomyolysis. A remarkable finding was galangin’s ability to enhance the endogenous antioxidant defence system. Treatment led to increased activity of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and glutathione reductase (GR), along with elevated levels of reduced glutathione (GSH). Concurrently, galangin treatment resulted in reduced malondialdehyde (MDA) and nitric oxide (NO) concentrations, demonstrating effective suppression of oxidative stress markers. Similarly, animals receiving galangin exhibited significantly reduced levels of pro-inflammatory mediators, including interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), myeloperoxidase (MPO), and nuclear factor kappa B (NF-κB). Simultaneously, there was an elevation in anti-inflammatory interleukin-10 (IL-10) compared to the untreated model group, in addition to a significant modulation of apoptotic pathways by decreased expression of pro-apoptotic proteins Bax and caspase-3, and increased in Bcl-2. The comprehensive findings from this study provide compelling evidence for galangin as a promising therapeutic agent against glycerol-induced acute kidney injury. Overall, galangin shows promise as a potential therapeutic agent for preventing or treating rhabdomyolysis-associated acute kidney injury.

Acute kidney injury (AKI) is a severe condition characterized by the rapid onset of kidney dysfunction within hours or days, leading to high rates of illness and death (Salem, 2021; Wen et al., 2023). Key causes of AKI include reduced kidney blood flow, structural damage to kidney tissues and blocked urine flow. Globally, AKI affects approximately 20% of patients (Li et al., 2017), with mortality rates around 20% in those without kidney damage, rising to 59% in AKI patients (Gonsalez et al., 2019). The condition disrupts the kidneys’ ability to filter waste and maintain fluid and electrolyte balance, leading to toxin buildup in the blood (Olano et al., 2024). AKI can also harm other organs, including the heart, brain and lungs. Although kidney function often recovers in survivors, AKI-related deaths remain high (Wu et al., 2017) and no effective treatment currently exists to prevent or manage the condition.
       
Among the multiple etiologies of AKI, rhabdomyolysis represents a major and well-recognized cause that significantly contributes to its incidence and severity. Rhabdomyolysis (RM), the breakdown of skeletal muscle releasing harmful substances like myoglobin into the bloodstream, is responsible for 10-40% of AKI cases and is a major contributor to its mortality (Stanley et al., 2024). Myoglobin causes kidney damage by constricting blood vessels, clogging kidney tubules and triggering oxidative stress, inflammation and cell death (Hebert et al., 2022). It also promotes the production of reactive radicals, activating nuclear factor kappa B (NF-κB), which increases inflammatory mediators like tumor necrosis factor-alpha (TNF-α) and interleukins during AKI. Antioxidants have shown promise in preventing or treating myoglobin-induced AKI (Al-Kharashi et al., 2023; El-khadragy et al., 2023).
       
Investigating plant-based therapeutics has emerged as a viable strategy for reducing the kidney damage linked to AKI, given the increased interest in natural substances as safer substitutes for synthetic medications. Alpinia officinarum (A. officinarum), a plant native to China and part of the Zingiberaceae family, is widely recognized for its medicinal properties due to its rich content of bioactive compounds like flavonoids, diarylheptanoids and volatile oils (Sun et al., 2023; Zheng et al., 2024; Wen et al., 2024). It contains unique isoflavones such as genistein and galangin, which offer various health benefits (Sharifi-Rad et al., 2021). Studies have shown that A. officinarum can protect kidney function in animal models (Javaid et al., 2021) and short-term use of its proteins has improved kidney function in patients with diabetes or chronic kidney disease (Aladaileh et al., 2021). Galangin, a key compound in A. officinarum, exhibits antioxidant, anti-inflammatory, antiapoptotic and anticancer properties (Guo et al., 2010). It has been shown to protect kidneys from cisplatin-induced damage in mice (Huang et al., 2017) and liver from injury caused by toxins (Luo et al., 2015). Additionally, galangin has been found to protect heart tissue from hypoxic-ischemic damage by activating the NRF2/HO-1 pathway (Yang et al., 2023).
               
While galangin’s kidney-protective effects have been documented, its role in counteracting glycerol-induced kidney injury and the underlying mechanisms involving NF-κB and apoptosis pathways remain unexplored. In this study, glycerol was used to induce AKI in rats to investigate galangin’s therapeutic potential. The study focused on evaluating key parameters and uncovering the mechanisms behind galangin’s protective effects, including its impact on oxidative stress, inflammation and apoptosis in kidney tissue.
Chemicals
 
Glycerol and galangin were procured from Sigma-Aldrich Chemical Co. (St. Louis, MO, USA). All other chemicals used in the study were of high analytical grade.
 
Experimental animals
 
Male Wistar albino rats were selected for this study due to their wide use in nephrotoxicity models and their availability in the animal facility of the Egyptian organization for biological products and vaccines (VACSERA). This ensured both scientific relevance and logistical accessibility, allowing for standardized handling and reliable replication of experimental procedures, the animals aged 3 months and weighing 180-200 g, were utilized in this study. The rats were sourced from VACSERA (Cairo, Egypt) and housed under controlled environmental conditions, including a 12-hour light-dark cycle, a temperature of 23± 2oC and relative humidity of 50±10%. They were provided with a standard diet and water ad libitum. Prior to the experiment, the rats were acclimatized for one week. All animal housing and experimental procedures were conducted in our institutional labs.
 
Induction of AKI and experimental design
 
Acute kidney injury (AKI) was induced by intramuscular injection of 50% glycerol (10 mL/kg, single dose) into the hind limbs of the rats after dilution in saline (0.9% NaCl). Rats were deprived of water for 24 hours before glycerol injection, as previously described (Kim et al., 2010).
       
Thirty-two rats were divided into four equal groups (n = 8 per group) as follows:

Group 1 (Negative Control) and was used as a baseline reference for all comparisons: Rats received an intramuscular injection of physiological saline (0.9% NaCl).
Group 2 (Gal): Rats were orally administered galangin at a dose of 100 mg/kg for 21 days, followed by an intramuscular injection of physiological saline (0.9% NaCl). The dosage of galangin was selected based on a previous study by Tomar et al., (2017).
Group 3 (AKI): Rats were injected with glycerol (50%, 10 mL/kg, intramuscular).
Group 4 (AKI + Gal): Rats were orally administered galangin at a dose of 100 mg/kg for 21 days, followed by an intramuscular injection of glycerol (50%, 10 mL/kg).
       
Twenty-four hours after glycerol administration, the rats were anesthetized via intraperitoneal injection of pentobarbital (100 mg/kg) and subsequently euthanized. Blood samples were collected from the retro-orbital plexus for biochemical analysis and the kidneys were immediately excised weighed and washed then homogenized in ice-cold 10 mM phosphate buffer (pH 7.4) to producea10%(w/v) homogenate for biochemical analysis. Using bovine serum albumin as a reference protein, the renal protein content was determined using the Lowry et al., (1951) technique.
 
Determination of kidney weight
 
The following mathematical formula was used to determine the relative kidney weight using the Almeer et al., (2019) method:


Assessment of the intensity of rhabdomyolysis
 
Using kits provided by Randox/Laboratory, Crumlin, UK, lactate dehydrogenase (LDH, Catalogue Number: LD3842, measuring ranges 8.8 - 635U/l) and creatine kinase (CK, Catalogue Number: CK3812, measuring ranges 9.16- 2886U/l)  were measured in accordance with the manufacturer’s procedure. At 2 to 8oC, reagents remain stable until they expire.
 
Determination of renal function biomarkers
 
Using kits (Randox/Laboratory, Crumlin, UK), serum levels of urea (Catalogue Number: CR2336, measuring range: 11.4–2460μmol/l) and creatinine (Catalogue Number: UR446, measuring range: 0.866–56.7mmol/l) were determined in accordance with the manufacturer’s instructions.
 
Estimation of renal non enzymatic oxidative stress markers
 
Using the Ohkawa et al., (1979) approach, lipid peroxidation was measured in terms of malondialdehyde (MDA). Furthermore, the Griess reagent was used to assess the amount of nitric oxide (NO) in renal tissues (Green et al., 1982). Additionally, Ellman (1959) formula for reduced glutathione (GSH) in renal tissue was used.
 
Measurement of kidney antioxidant enzymatic activities
 
The enzymes catalase (CAT) and superoxide dismutase (SOD) were tested in accordance with Aebi (1984) and Nishikimi et al., (1972), respectively. Additionally, glutathione reductase (GR) was calculated in accordance with De Vega et al., (2002) and glutathione peroxidase (GPx) was evaluated in accordance with Paglia and Valentine (1967).
 
Assessment of inflammatory related markers
 
TNF-α (Catalogue Number: NBP1-92681), interleukin-1β (IL-1β; Catalogue Number: NBP1-92702), interleukin-10 (IL-10; Catalogue Number: NBP1-92701) and NF-κB (Catalogue Number: NB100-2176) were measured in accordance with the manufacturer’s instructions to assess the renal inflammatory response (Novus Biologicals, Centennial, CO, USA). Myeloperoxidase (MPO) activity was measured using the Bradley et al., (1982) methodology.

Assessment of the renal apoptotic markers
 
The levels of apoptotic proteins (Bax, caspase-3 and Bcl- 2) in renal tissues of all tested groups were measured by ELISA kit (Cusabio (Wuhan, China)) following the manufacturer’s information.
 
Statistical analysis
 
Data analysis was done using the Statistical Package for the Social Sciences (SPSS). The mean±standard error of the mean (SEM) was used to represent the results. The significance was assessed using Duncan’s test after one-way analysis of variance (ANOVA).  At p<0.05, the acceptable threshold of significance was determined.
The results of present study demonstrate the protective effects of galangin on renal function and oxidative stress in a rat model of acute kidney injury (AKI) induced by glycerol. Acute kidney injury (AKI) is marked by a rapid decline in glomerular filtration rate (GFR) and the accumulation of nitrogenous waste in the bloodstream (Zywno et al., 2024). Although nephrotoxic agents like gentamicin and cisplatin are commonly used to model AKI in animals, they do not fully capture the pathophysiology of AKI resulting from rhabdomyolysis (RM). RM, a significant contributor to AKI, occurs due to the breakdown of skeletal muscle, leading to the release of muscle enzymes and proteins into circulation (Wang et al., 2021). As illustrated in Fig 1, the relative kidney weight in the AKI group was significantly higher (1.31±0.05) compared to the control group (0.71±0.08) at P<0.05, indicating renal damage. Elevated plasma creatinine and urea levels further confirmed impaired kidney function in the AKI group. However, treatment with galangin significantly improved renal function biomarkers, suggesting its nephroprotective potential (Fig 1).

Fig 1: Impact of galangin pretreatment on kidney weight, urea and creatinine levels in a glycerol-induced acute kidney injury (AKI) model in rats.


       
In this study, a glycerol-induced RM model was employed to replicate key clinical features of AKI, including myoglobin accumulation, tubular obstruction and renal impairment. The administration of galangin demonstrated notable protective effects against RM-induced AKI, as reflected in functional and biochemical improvements. Additionally, serum creatine kinase (CK) (203.38±15.21) and lactate dehydrogenase (LDH) (38.83±7.23) levels, which were markedly increased (P<0.05) in the AKI group as a result of glycerol intramuscular injection. These elevations were significantly reduced by galangin administration at a dose of 100 mg/kg (113.59±10.45) and (21.10±35) respectively, at P<0.05 (Fig 2). These findings highlight galangin’s ability to mitigate muscle fibre damage caused by glycerol injection. AKI following RM is characterized by elevated creatine kinase (CK) and lactate dehydrogenase (LDH) levels, indicating muscle damage, in agreement with previous studies (Yin and Wang, 2024). Glycerol-induced RM triggers myoglobin release, which, when exceeding plasma protein binding capacity, accumulates in renal tubules, leading to obstruction and tubular cell death (Qiao et al., 2023). The study observed increased kidney weight, along with elevated serum creatinine and urea levels, which are indicative of renal edema and impaired kidney function, consistent with earlier findings (Brookes and Power, 2022; El-Khadragy et al., 2024).

Fig 2: Effect of galangin pretreatment on the rhabdomyolysis-related biomarkers in glycerol-induced acute kidney injury (AKI) model in rats.


       
The antioxidant enzymatic activity in renal tissues was evaluated, as shown in Fig 3. Rats treated with glycerol exhibited significantly reduced (P<0.05) activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) and glutathione reductase (GR) compared to the sham group. In contrast, pretreatment with galangin significantly enhanced (P<0.05) the activities of these enzymes, with SOD and GR levels in the galangin-treated group reaching levels comparable to those of the control group. Furthermore, non-enzymatic oxidant biomarkers were analyzed (Fig 4). Glycerol administration led to a significant decline (P<0.05) in glutathione (GSH) levels and a concurrent increase (P<0.05) in malondialdehyde (MDA) and nitric oxide (NO) levels compared to the sham group. Galangin treatment reversed these changes, demonstrating its ability to alleviate oxidative stress in renal tissues by enhancing antioxidant defenses and reducing oxidant markers.

Fig 3: Effect of galangin pretreatment on renal enzymatic antioxidant biomarkers (SOD, CAT, GPx and GR) in glycerol-induced acute kidney injury (AKI) model in rats.



Fig 4: Effect of galangin pretreatment on GSA, NO and MDA in glycerol-induced acute kidney injury (AKI) model in rats.


       
The anti-inflammatory effects of galangin were also investigated (Fig 5). Glycerol-induced AKI was associated with a significant increase (P<0.05) in pro-inflammatory cytokines (IL-1β, TNF-α and MPO) and a decrease (P<0.05) in the anti-inflammatory cytokine IL-10. Additionally, nuclear factor-kappa B (NF-κB) levels were elevated (P<0.05) in the AKI group compared to the sham group. Remarkably, galangin administration at 100 mg/kg restored the levels of these inflammatory markers significantly as compared to AKI group. Oxidative stress, primarily driven by reactive oxygen species (ROS), is a key factor in the development of glycerol-induced AKI (Hebert et al., 2022). Myoglobin and heme derivatives released during RM contribute to excessive ROS production, which leads to oxidative damage in renal tissues. In this study, glycerol administration was associated with increased levels of malondialdehyde (MDA) and nitric oxide (NO), along with a reduction in glutathione (GSH) and decreased activity of key antioxidant enzymes (SOD, CAT, GPx and GR). These results are in line with previous research (Gyurászová et al., 2020; Salem et al., 2023).

Fig 5: Effect of galangin pretreatment on inflammatory biomarkers in glycerol-induced acute kidney injury (AKI) model in rats.


       
Galangin, a flavonoid compound found in Alpinia officinarum improves kidney function in acute kidney injury (AKI) by reducing oxidative stress, thereby preserving glomerular filtration rate (GFR) and enhancing creatinine and urea clearance as observed in the present results (Alsawaf et al., 2022). Its antioxidant properties - due to its phenolic structure - neutralize reactive oxygen species (ROS), preventing lipid peroxidation, mitochondrial damage and endothelial dysfunction (Aloud et al., 2017). By upregulating the Nrf2/HO-1 pathway, galangin boosts antioxidant defenses, enhances glutathione (GSH) levels and protects renal tubular cells. It also inhibits the NF-κB pathway, reducing inflammation and cytokine production, which helps maintain kidney structure and prevent fibrosis (Khawaja et al., 2024; Almohawes et al., 2025). Additionally, galangin restores nitric oxide (NO) bioavailability, improving renal blood flow and preventing vasoconstriction-induced damage (Ahmad et al., 2018). By protecting renal cells from oxidative and inflammatory injury, galangin preserves glomerular and tubular integrity, ensuring efficient creatinine and urea excretion, ultimately mitigating AKI progression.
       
The present study examined apoptotic markers in renal tissues (Fig 6). Glycerol injection resulted in increased (P<0.05) expression of pro-apoptotic proteins (Bax and caspase-3) and decreased (P<0.05) levels of the anti-apoptotic protein Bcl-2 markedly compared to the sham group. Galangin co-administration effectively suppressed apoptosis in renal tissues, as evidenced by the amelioration of these markers significantly. Inflammation and leukocyte infiltration are characteristic features of RM-induced AKI. Excess ROS production amplifies inflammation by activating the NF-kB pathway, which regulates the expression of pro-inflammatory cytokines such as IL-1β, TNF-α and MPO (McSweeney et al., 2021). The current study observed a significant increase in these inflammatory markers following glycerol injection, this is in agreement with previous studied. MPO, released by neutrophils, generates hypochlorous acid, which contributes to lipid peroxidation and oxidative stress, further exacerbating renal damage (Kisic et al., 2016).

Fig 6: Effect of galangin pretreatment on Apoptotic biomarkers in glycerol-induced acute kidney injury (AKI) model in rats.


       
Galangin’s anti-inflammatory effects in acute kidney injury (AKI) are primarily driven by its polyphenolic structure, rich in hydroxyl (-OH) groups, which enable ROS scavenging and modulation of key inflammatory pathways (Ashkar et al., 2022). It inhibits NF-κB signaling, reducing the expression of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, COX-2) and suppressing neutrophil infiltration. Additionally, galangin activates the Nrf2/HO-1 pathway, enhancing heme oxygenase-1 (HO-1) production to counteract oxidative stress and inflammation. It also inhibits iNOS, preventing excessive nitric oxide (NO) production and suppresses the MAPK pathway (p38, JNK, ERK), further reducing cytokine-driven inflammation (Khawaja et al., 2024).
       
The interplay between oxidative stress and inflammation promotes apoptotic cell death in AKI. In this study, glycerol administration led to increased expression of pro-apoptotic markers, including Bax and caspase-3, while reducing the levels of the anti-apoptotic protein Bcl-2. These observations suggest an apoptotic shift, consistent with previous reports (Guttà et al., 2020). Bcl-2 and Bax regulate mitochondrial membrane integrity, with Bcl-2 favoring cell survival and Bax promoting apoptosis (Qiao et al., 2023).
       
Galangin attenuates apoptosis in acute kidney injury (AKI) by regulating key pro-apoptotic and anti-apoptotic markers. Its polyphenolic structure enables it to neutralize oxidative stress and inhibit inflammation, both of which contribute to apoptosis (Aladaileh et al., 2019). Galangin downregulates pro-apoptotic proteins such as Bax and caspase-3, preventing mitochondrial dysfunction and cell death. Simultaneously, it upregulates anti-apoptotic Bcl-2, preserving mitochondrial integrity and promoting cell survival. Additionally, galangin activates the PI3K/Akt pathway, which enhances cell survival by suppressing apoptotic signaling (Palachai et al., 2025). By reducing oxidative damage, inhibiting NF-κB-mediated inflammation and balancing apoptotic regulators, galangin effectively protects renal cells from programmed cell death, aiding in kidney recovery during AKI.
               
Although this study highlights the renoprotective effects of galangin, its clinical applicability is limited by the pretreatment duration of 21 days. Future research should explore the efficacy of galangin administered post-AKI induction or simultaneously with injury onset to better mimic clinical conditions. Additionally, further investigation is warranted to delineate the precise molecular mechanisms through which galangin modulates oxidative stress, inflammation and apoptosis in AKI.
In conclusion, galangin exerts protective effects against glycerol-induced AKI through multiple mechanisms, including the enhancement of antioxidant defenses, reduction of oxidative stress, suppression of inflammation and inhibition of apoptosis. These findings underscore the therapeutic potential of galangin in the management of AKI and provide a foundation for further research into its clinical applications.
This study was supported by Princess Nourah bint Abdulrahman University Researchers Supporting Project number (PNURSP2025R23), Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia and also was supported by Researchers Supporting Project (RSP2025R25), King Saud University, Riyadh, Saudi Arabia.
 
Disclaimers
 
The views and conclusions expressed in this article are solely of the authors and do not necessarily represent the views of our affiliated institutions. The authors are responsible for the accuracy and completeness of the information provided, but do not accept any liability for any direct or indirect losses resulting from the use of this content.
 
Informed consent
 
All animal procedures of these experiments were approved by the Committee of Experimental Animal Care and handling techniques were approved by the University of Animal Care Committee. By the National Institutes of Health (NIH) Guidelines for the Care and Use of Laboratory Animals, 8th edition, all protocols and animal handling at the Department of Zoology, Faculty of Science, Helwan University were approved by the Committee on Research Ethics for Laboratory Animal Care (Cairo, Egypt; approval no. HU2021/Z/RKA0921-01). 
There are no conflicts of interest regarding the publication of this article. No funding or sponsorship influenced the design of the study, data collection, analysis, decision to publish, or preparation of the manuscript.

  1. Aebi, H. (1984). Catalase in vitro. Methods Enzymology. 105: 121-126.

  2. Ahmad, A., Dempsey, S.K., Daneva, Z., Azam. M., Li, N., Li, PL., Ritter, J.K. (2018). Role of nitric oxide in the cardiovascular and renal systems. International Journal of Molecular Science. 3. 19(9): 2605. doi: 10.3390/ijms19092605. PMID: 30177600; PMCID: PMC6164974.

  3. Aladaileh, S.H., Abukhalil, M.H., Saghir, S.A.M., Hanieh, H., Alfwuaires, M.A., Almaiman, A.A., Bin-Jumah, M., Mahmoud, A.M. (2019). Galangin activates Nrf2 signaling and attenuates oxidative damage, inflammation and apoptosis in a rat model of cyclophosphamide-induced hnepatotoxicity. Biomolecules. 5. 9(8): 346. doi: 10.3390/biom9080346. PMID: 31387329; PMCID: PMC6723184.

  4. Aladaileh, S.H., Al-Swailmi, F.K., Abukhalil, M.H., Shalayel, M.H. (2021). Galangin protects against oxidative damage and attenuates inflammation and apoptosis via modulation of NF-κB p65 and caspase-3 signaling molecules in a rat model of diabetic nephropathy. Journal of Physiology and Pharmacology. 72(1). doi: 10.26402/jpp.2021.1.04. Epub 2021 Jun 3. PMID: 34099583.

  5. Al-Kharashi, L., Attia, H., Alsaffi, A., Almasri, T., Arafa, M., Hasan, I., Alajami, H., Ali, R., Badr, A. (2023). Pentoxifylline and thiamine ameliorate rhabdomyolysis-induced acute kidney injury in rats via suppressing TLR4/NF-κB and NLRP-3/caspase- 1/gasdermin mediated-pyroptosis. Toxicol Appl Pharmacol. 15(461): 116387. doi: 10.1016/j.taap.2023.116387. Epub 2023 Jan 20. PMID: 36690085.

  6. Almeer, R.S., AlBasher, G.I., Alarifi, S., Alkahtani, S., Ali, D., Abdel Moneim, A.E. (2019). Royal jelly attenuates cadmium-induced nephrotoxicity in male mice. Scientific Report. 9: 5825.

  7. Almohawes, Z.N., El-Khadragy, M.F., Al-Megrin, W.A.I. and Salem, F.E.H. (2025). Potential neuromodulatory effects of prodigiosin- conjugated silver nanoparticles: Implications for epilepsy management and their impact on obesity in rat model. Indian Journal of Animal Research. 59(6): 953-962. doi: 10. 18805/IJAR.BF-1915. 

  8. Aloud, A.A., Veeramani, C., Govindasamy, C., Alsaif, M.A., Al-Numair, K.S. (2017). Galangin, a natural flavonoid reduces mitochondrial oxidative damage in streptozotocin-induced diabetic rats. Redox Report. 23(1): 29-34. doi: 10.1080/13510002.2017. 1365224. Epub 2017 Aug 16. PMID: 28813209; PMCID: PMC6748697.

  9. Alsawaf, S., Alnuaimi, F., Afzal, S., Thomas, R.M., Chelakkot, A.L., Ramadan, W.S., Hodeify, R., Matar, R., Merheb, M., Siddiqui, S.S., Vazhappilly, C.G. (2022). Plant flavonoids on oxidative stress-mediated kidney inflammation. Biology (Basel). 11(12): 1717. doi: 10.3390/biology11121717. PMID: 36552226; PMCID: PMC9774981.

  10. Ashkar, F., Bhullar, K.S., Wu, J. (2022). The effect of polyphenols on kidney disease: Targeting mitochondria. Nutrients. 14(15): 3115. doi: 10.3390/nu14153115. PMID: 35956292; PMCID: PMC9370485.

  11. Bradley, P.P., Priebat, D.A., Christensen, R.D., Rothstein, G. (1982). Measure- ment of cutaneous inflammation: Estimation of neutrophil content with an enzyme marker. Journal of Investigative Dermatology. 78: 206-209.

  12. Brookes, E.M. and Power, D.A. (2022). Elevated serum urea-to-creatinine ratio is associated with adverse inpatient clinical outcomes in non-end stage chronic kidney disease. Sci Rep. 2022 Dec 2. 12(1): 20827. doi: 10.1038/s41598-022-25254-7. PMID: 36460694; PMCID: PMC9718835.

  13. De Vega, L., Fernandez, R.P., Mateo, M.C., Bustamante, J.B., Herrero, A.M., Munguira, E.B. (2002). Glutathione determination and a study of the activity of glutathione-peroxidase, glutathione-transferase and glutathione-reductase in renal transplants. Renal Feiler. 24: 421-432.

  14. El-Khadragy, M.F., AlDurgham, N.M., Hamed, S.S., Salem, F.E.H., Aljumaa, M.A., Yehia, H.M. and Alkhateeb, M.A. (2024). Immunomo- dulatory effect of Eugenia caryophyllata against Vitamin D-induced Toxicity in Liver of Male Westar Rats. Indian Journal of Animal Research. 58(2): 200-209. doi: 10. 18805/IJAR.BF-1711.

  15. El-khadragy, M.F., Salem, F.E.H., Yehia, H.M. and Al-Megrin, W.A. (2023). Therapeutic potency of prodigiosin conjugated with silver-nanoparticles in male rats exposed to cadmium chloride- induced testicular toxicity. Indian Journal of Animal Research. 57(10): 1322-1329. doi: 10.18805/IJAR.BF-1663. 

  16. Ellman, G.L. (1959). Tissue sulfhydryl groups. Archives of Biochemistry and Biophysics. 82: 70-77.

  17. Gonsalez, S.R., Cortês, A.L., Silva, R.C.D., Lowe, J., Prieto, M.C., Silva Lara, L.D. (2019) Acute kidney injury overview: From basic findings to new prevention and therapy strategies. Pharmacology and Therapeutics. 200:1-12. doi: 10.1016/ j.pharmthera.2019.04.001. Epub 2019 Apr 6. PMID: 30959059; PMCID: PMC10134404.

  18. Green, L.C., Wagner, D.A., Glogowski, J., Skipper, P.L., Wishnok, J.S., Tannenbaum, S.R. (1982). Analysis of nitrate, nitrite and [15N] nitrate in biological fluids. Analytical Biochemistry. 126: 131-138

  19. Guo, A.J., Xie, H.Q., Choi, R.C., Zheng, K.Y., Bi, C.W., Xu, S.L., Dong, T.T., Tsim, K.W. (2010). Galangin, a flavonol derived from rhizoma alpiniae officinarum, inhibits acetylcholinesterase activity in vitro. Chemico-Biological Interactions. 187(1-3): 246-248. doi: 10.1016/j.cbi.2010.05.002. Epub 2010 May 7. PMID: 20452337.

  20. Guttà, C., Rahman, A., Aura, C., Dynoodt, P., Charles, E.M., Hirschenhahn, E., Joseph, J., Wouters, J., de Chaumont, C., Rafferty, M., Warren, M., van den Oord, J.J., Gallagher, W.M., Rehm, M. (2020). Low expression of pro-apoptotic proteins Bax, Bak and Smac indicates prolonged progression-free survival in chemotherapy-treated metastatic melanoma. Cell Death and Disease. 11(2): 124. doi: 10.1038/s41419-020-2309-3. PMID: 32054850; PMCID: PMC7018795.

  21. Gyurászová, M., Gurecká, R., Bábíèková, J., Tóthová, Ľ. (2020). oxidative stress in the pathophysiology of kidney disease: implications for noninvasive monitoring and identification of biomarkers. Oxidative Medicine and Cellular Longevity. 2020: 5478708. doi: 10.1155/2020/5478708. PMID:3208- 2479; PMCID: PMC7007944.

  22. Hebert, J.F., Burfeind, K.G., Malinoski, D., Hutchens, M.P. (2022). Molecular mechanisms of rhabdomyolysis-induced kidney injury: From bench to bedside. Kidney International Reports. 8(1): 17-29. doi: 10.1016/j.ekir.2022.09.026. PMID: 36644345; PMCID: PMC9831947.

  23. Huang, Y.C., Tsai, M.S., Hsieh, P.C., Shih, J.H., Wang, T.S., Wang, Y.C., Lin, T.H., Wang, S.H. (2017). Galangin ameliorates cisplatin- induced nephrotoxicity by attenuating oxidative stress, inflammation and cell death in mice through inhibition of ERK and NF-kappaB signaling. Toxicology and Applied Pharmacology. 329: 128-139. doi: 10.1016/j.taap.2017.05. 034. Epub 2017 May 27. PMID: 28558962.

  24. Javaid, F., Mehmood, M.H., Shaukat, B. (2021). Hydroethanolic extract of A. officinarum hance ameliorates hypertension and causes diuresis in obesogenic feed-fed rat model. Frontiers in Pharmacology. 12: 670433. doi: 10.3389/fphar.2021. 670433. PMID: 34305591; PMCID: PMC8299705.

  25. Khawaja, G., El-Orfali, Y., Shoujaa, A., Abou Najem, S. (2024). Galangin: A promising flavonoid for the treatment of rheumatoid arthritis- mechanisms, evidence and therapeutic potential. Pharma- ceuticals (Basel). 17(7): 963. doi: 10.3390/ph17070963. PMID: 39065811; PMCID: PMC11279697.

  26. Kim, J.H., Lee, S.S., Jung, M.H., Yeo, H.D., Kim, H.J., Yang, J.I., Roh, G.S., Chang, S.H., Park, D.J. (2010). N-acetylcysteine attenuates glycerol- induced acute kidney injury by regulating MAPKs and Bcl-2 family proteins. Nephrol, Dial, Transplant : Off Publ Europ Dial Transplant Assoc - European Renal Association. 25:1435-1443.

  27. Kisic, B., Miric, D., Dragojevic, I., Rasic, J., Popovic, L. (2016). Role of myeloperoxidase in patients with chronic kidney disease. oxidative medicine and cellular longevity. 2016: 1069743. doi: 10.1155/2016/1069743. Epub 2016 Apr 3. PMID: 27127544; PMCID: PMC4834151.

  28. Li, X., Wang, J., Zhang, H., Zhang, Q. (2017). Renoprotective effect of low- molecular-weight sulfated polysaccharide from the seaweed Lami- naria japonica on glycerol-induced acute kidney injury in rats. International Journal of Biological Macromolecules. 95: 132-137.

  29. Lowry, O.H., Rosebrough, N.J., Farr, A.L., Randall, R.J. (1951). Protein measurement with the Folin phenol reagent. Journal of Biological Chemistry. 193: 265-275.

  30. Luo, Q., Zhu, L., Ding, J., Zhuang, X., Xu, L., Chen, F. (2015). Protective effect of galangin in Concanavalin A-induced hepatitis in mice. Drug Design, Development and Therapy. 9: 2983- 2992. doi: 10.2147/DDDT.S80979. PMID: 26089647; PMCID: PMC4468934.

  31. McSweeney, K.R., Gadanec, L.K., Qaradakhi, T., Ali, B.A., Zulli, A., Apostolopoulos, V. (2021). Mechanisms of cisplatin- induced acute kidney injury: Pathological mechanisms, pharmacological interventions and genetic mitigations. Cancers (Basel). 13(7): 1572. doi: 10.3390/cancers13071- 572. PMID: 33805488; PMCID: PMC8036620.

  32. Nishikimi, M., Appaji, N., Yagi, K. (1972). The occurrence of superox- ide anion in the reaction of reduced phenazine methosulfate and molecular oxygen. Biochemical and Biophysical Research Communications. 46: 849-854.

  33. Ohkawa, H., Ohishi, N., Yagi, K. (1979). Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem. 95: 351-358. 

  34. Olano, C.G., Akram, S.M., Hashmi, M.F., Bhatt, H. (2024). Uremic Encephalopathy. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan–. PMID: 33231997.

  35. Paglia, D.E. and Valentine, W.N. (1967). Studies on the quantitative and quali- tative characterization of erythrocyte glutathione peroxi- dase. Journal of Laboratory and Clinical Medicine. 70: 158-169.

  36. Palachai, N., Supawat, A., Kongsui, R., Klimaschewski, L., Jittiwat, J. (2025). Galangin’s neuroprotective role: Targeting oxidative stress, inflammation and apoptosis in ischemic stroke in a rat model of permanent middle cerebral artery occlusion. International Journal of Molecular Sciences. 26(5): 1847. doi: 10.3390/ijms26051847. PMID: 40076473; PMCID: PMC11899015.

  37. Qiao, O., Wang, X., Wang, Y., Li, N., Gong, Y. (2023). Ferroptosis in acute kidney injury following crush syndrome: A novel target for treatment. Journal of Advanced Research. 54: 211-222. doi: 10.1016/j.jare.2023.01.016. Epub 2023 Jan 24. PMID: 36702249; PMCID: PMC10703611.

  38. Salem FH (2021). Syzygium cumini (L) extract ameliorates aluminium chloride-induced acute hepatic and renal toxicity in rats. Egyptian Academic Journal of Biological Sciences C. Physiology and Molecular Biology. 13(2): 73-86.

  39. Salem, F.E.H., Abdel-Gaber, R., Al-Megrin, W.A. and El-Khadragy, M.F. (2023). Therapeutic potency of camellia sinensis extract on neurochemical and oxidative changes correlates to autistic disorder in rat pups model. Indian Journal of Animal Research. 57(12): 1652-1661. doi: 10.18805/IJAR.BF-1666. 

  40. Sharifi-Rad, J., Quispe, C., Imran, M., Rauf, A., Nadeem, M., Gondal, T.A., Ahmad, B., Atif, M., Mubarak, M.S., Sytar, O., Zhilina, O.M., Garsiya, E.R., Smeriglio, A., Trombetta, D., Pons, D.G., Martorell, M., Cardoso, S.M., Razis, A.F.A., Sunusi, U., Kamal, R.M., Rotariu, L.S., Butnariu, M., Docea, A.O., Calina, D. (2021). Genistein: An Integrative Overview of Its Mode of Action, Pharmacological Properties and Health Benefits. Oxidative Medicine and Cellular Longevity. 2021:3268136. doi: 10.1155/2021/3268136. PMID: 34336089; PMCID: PMC8315847.

  41. Stanley, M., Chippa, V., Aeddula, N.R., Quintanilla Rodriguez, B.S., Adigun, R. (2024). Rhabdomyolysis. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan–. PMID: 28846335.

  42. Sun, Y. Y., Feng, J., Wei, J. H., Zhan, Z. L., Liu, Y. Y. (2023). Herbal textual research on alpiniae officinarum rhizoma in famous classical formulas. Chinese Journal of Experimental Traditional Medical Formulae. 29: 94-103.

  43. Tomar, A., Vasisth, S., Khan, SI., Malik, S., Nag, T.C., Arya, D.S., Bhatia, J. (2017). Galangin ameliorates cisplatin induced nephrotoxicity in vivo by modulation of oxidative stress, apoptosis and inflammation through interplay of MAPK signaling cascade. Phytomedicine. 34:154-161. doi: 10.1016/j.phymed.2017.05. 007. Epub 2017 Jun 15. PMID: 28899498.

  44. Wang, S.Y., Zhang, C.Y., Cai, G.Y., Chen, X.M. (2021). Method used to establish a large animal model of drug-induced acute kidney injury. Experimental Biology and Medicine (Maywood). 246(8): 986-995. doi: 10.1177/1535370220981756. Epub 2021 Jan 19. PMID: 33467911; PMCID: PMC8024500.

  45. Wen, H., Kuang, Y., Lian, X., Li, H., Zhou, M., Tan, Y., et al. (2024). Physicochemical characterization, antioxidant and anticancer activity evaluation of an acidic polysaccharide from alpinia officinarum hance. Molecules. 29: 1810. 10.3390/molecules 29081810.

  46. Wen, Y., Xu, L., Melchinger, I., Thiessen-Philbrook, H., Moledina, D.G., Coca, S.G., Hsu, C.Y., Go, A.S., Liu, K.D., Siew, E.D., Ikizler, T.A., Chinchilli, V.M., Kaufman, J.S., Kimmel, P.L., Himmelfarb, J., Cantley, L.G., Parikh, C.R. (2023). ASSESS-AKI consortium. longitudinal biomarkers and kidney disease progression after acute kidney injury. JCI Insight. 8(9): e167731. doi: 10. 1172/jci.insight.167731. PMID: 36951957; PMCID:PMC10 243801.

  47. Wu, J., Pan, X., Fu, H., Zheng, Y., Dai, Y., Yin, Y., Chen, Q., Hao, Q., Bao, D., Hou, D. (2017). Effect of curcumin on glycerol-induced acute kidney injury in rats. Scientific Report. 7: 1-11

  48. Yang, T., Liu, H., Yang, C., Mo, H., Wang, X., Song, X., Jiang, L., Deng, P., Chen, R., Wu, P., Chen, A., Yan, J. (2023). Galangin attenuates myocardial ischemic reperfusion-induced ferroptosis by Targeting Nrf2/Gpx4 Signaling Pathway. Drug Design, Development and Therapy. 17: 2495-2511. doi: 10.2147/DDDT.S409232. PMID: 37637264; PMCID: PMC10460190.

  49. Yin, X. and Wang, W. (2024). Predictive value of serum myoglobin and lactate dehydrogenase in rhabdomyolysis-induced acute kidney injury from severe heatstroke. American Journal of Translational Research. 16(4): 1477-1483. doi: 10.62347/QSRI4858. PMID: 38715805; PMCID: PMC11070377.

  50. Zheng, X., Zhang, Y., Tan, Y., Li, Y., Xue, Q., Li, H., et al. (2024). Alpinia officinarum Hance extract ameliorates diabetic gastroparesis by regulating SCF/c-kit signaling pathway and rebalancing gut microbiota. Fitoterapia. 172: 105730. 10.1016/j.fitote. 2023.105730.

  51. Zywno, H., Figiel, W., Grat, M., Nazarewski, S., Galazka, Z., Malyszko, J. (2024). Can novel biomarkers effectively predict acute kidney injury in liver or kidney transplant recipients? international Journal of Molecular Sciences. 25(22): 12072. doi: 10.3390/ijms252212072. PMID: 39596140; PMCID: PMC11593440.

Editorial Board

View all (0)