Centhaquine Restores Renal Blood Flow and Protects Tissue Damage After Hemorrhagic Shock and Renal Ischemia

Amaresh K Ranjan, Zhong Zhang, Seema Briyal, Anil Gulati, Amaresh K Ranjan, Zhong Zhang, Seema Briyal, Anil Gulati

Abstract

Background: Centhaquine (CQ) (Lyfaquin®) is in late stage clinical development as a safe and effective first-in-class resuscitative agent for hemorrhagic shock patients (NCT02408731, NCT04056065, and NCT04045327). Acute kidney injury (AKI) is known to be associated with hemorrhagic shock. Hence, effect of CQ on protection of kidneys from damage due to hemorrhagic shock was investigated. Methods: To assess effect of CQ on AKI in shock, we created a rat model with hemorrhagic shock and AKI. Renal arteries were clamped and de-clamped to induce AKI like ischemia/reperfusion model and hemorrhage was carried out by withdrawing blood for 30 min. Rats were resuscitated with CQ (0.02 mg/kg) for 10 min. MAP, heart rate (HR), and renal blood flow (RBF) were monitored for 120 min. Results: CQ produced a significant improvement in RBF compared to vehicle (p< 0.003) even though MAP and HR was similar in CQ and vehicle groups. Blood lactate level was lower (p = 0.0064) in CQ than vehicle at 120 min post-resuscitation. Histopathological analysis of tissues indicated greater renal damage in vehicle than CQ. Western blots showed higher HIF-1α (p = 0.0152) and lower NGAL (p = 0.01626) levels in CQ vs vehicle. Immunofluorescence in the kidney cortex and medulla showed significantly higher (p< 0.045) expression of HIF-1α and lower expression of Bax (p< 0.044) in CQ. Expression of PHD 3 (p< 0.0001) was higher, while the expression of Cytochrome C (p = 0.01429) was lower in the cortex of CQ than vehicle. Conclusion: Results show CQ (Lyfaquin®) increased renal blood flow, augmented hypoxia response, decreased tissue damage and apoptosis following hemorrhagic shock induced AKI, and may be explored to prevent/treat AKI. Translational Statement: Centhaquine (CQ) is safe for human use and currently in late stage clinical development as a first-in-class resuscitative agent to treat hemorrhagic shock. In the current study, we have explored a novel role of CQ in protection from hemorrhagic shock induced AKI, indicating its potential to treat/prevent AKI.

Keywords: acute kidney injury; centhaquine; hemorrhage; hypoxia inducible factors; resuscitation; shock.

Conflict of interest statement

AG is an employee of Pharmazz, Inc., and has issued and pending patents related to this study. Midwestern University is the patent assignee with AG as an inventor of this technology, while Pharmazz Inc. holds its exclusive worldwide license and is engaged in the clinical development and commercialization of centhaquine (CQ) for human use. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Ranjan, Zhang, Briyal and Gulati.

Figures

FIGURE 1
FIGURE 1
Schematic representation of in vivo experiment in rats involving hypovolemic shock, acute kidney injury with kidney ischemia/reperfusion (I/R) and resuscitation with CQ. Procedure from anesthesia to the end of the in vivo experiment is shown with brief description of each step and time intervals of some important steps. B.G.–indicates a small volume blood collection for analyzing blood gas components.
FIGURE 2
FIGURE 2
Effect of CQ on kidney blood flow in a rat model of kidney ischemia/reperfusion with hypovolemic shock. (A), 1 and 2, representative images of blood flow in kidneys of vehicle 1) and CQ 2). Upper panel in 1 and 2 shows kidney images obtained from PeriCam PSI blood flow imaging system. Red represents highest blood flow and blue represents the lowest. The lower panels in 1 and 2 show the blood flow graphs–Blue line indicates blood flow in complete imaged area, Black line indicates blood flow in the kidney. N = 7 for vehicle and N = 8 for CQ. (B), a graphical representation of kidney blood flow at different time points (1–baseline, 2–after hemorrhage, 3–after de-clamping renal arteries, 4–0 min post-perfusion, 5–30 min post-perfusion, 6–90 min post-perfusion and 7–120 min post-perfusion). Data represents mean ± SEM values. (C), the graph represents percent change in blood lactate levels in CQ and vehicle rats at different time points–after hemorrhage (0 min of resuscitation), at 30 and 120 min of resuscitation. Baseline is indicated with dotted lines. Data represents mean ± SEM values. N = 7 for vehicle and N = 8 for CQ. Statistical analysis–unpaired t-tests.
FIGURE 3
FIGURE 3
Effect of CQ on cardiovascular activities, HR and MAP. (A, B), a graphical representation of mean HR (A) and MAP (B) at different time points (1–baseline, 2–after hemorrhage, 3–after de-clamping renal arteries, 4–0 min post-perfusion, 5–30 min post perfusion, 6–90 min post-perfusion and 7–120 min post-perfusion). Data represents mean ± SEM values. N = 7 for vehicle and N = 8 for CQ. Statistical analysis–unpaired t-tests.
FIGURE 4
FIGURE 4
Effect of CQ on expression of hypoxia responsive factors in kidneys. A and B, representative western blots of HIF-1α (A) and HIF-1β (B) and their respective GAPDH (loading control). (C, D), graphical representation of HIF-1α/GAPDH (C) and HIF-1β/GAPDH (D) normalized data. The error bar represents mean ± SEM. N = 4. Black vertical lines in A and B show that the bands were cropped from the same blot (full blots are provided as Supplementary Figures S1, S2). Statistical analysis Ordinary One Way ANOVA and Fisher’s test. E and F, representative images of immunofluorescence of HIF-1α (red) and HIF-1β (green) in cortex (E) and medulla (F) in sham, vehicle (control) and CQ. (G–J), graphs of mean fluorescence intensity of HIF-1α in cortex (G) and medulla (H), and of HIF-1β in cortex (I) and medulla (J). N = 4. Nuclei in all immunofluorescence microscopy images were stained with DAPI (blue). Statistical analysis- Ordinary One Way ANOVA and Fisher’s test.
FIGURE 5
FIGURE 5
Expression of hypoxia responsive factor, PHD 3 in kidney cortex. (A), representative images of immunofluorescence of PHD 3 (red) in cortex (A) in sham, vehicle (control) and CQ. (B), a graph of mean fluorescence intensity of PHD 3 in cortex. N = 4. Nuclei in all immunofluorescence microscopy images were stained with DAPI (blue). Statistical analysis Ordinary One Way ANOVA and Fisher’s test.
FIGURE 6
FIGURE 6
Effect of CQ on kidney tissue damage. (A), representative microscopy images of H and E stained kidney tissue sections from sham, saline and CQ treated rats. Some of the dilated cortical tubules are marked with “black arrows”, vacuoles (clear spaces) in tubules are shown by “blue arrow heads”, a highly vacuolated tubule is “encircled with blue line”, necrotic cells are indicated by “thin black arrows” and tubules with hyaline casts are shown with “white arrows”. Bar scale = 50 µm. (B), a graph showing pathological damage score of the kidney tissues in sham, vehicle and CQ treated rats, Calculated on the basis of scores of tubular dilatation (C), hyaline casts (D) and tubular necrosis or degeneration (E). Error bars represent mean ± SEM. N = 4. Statistical analysis- Ordinary One Way ANOVA and Fisher’s test.
FIGURE 7
FIGURE 7
Effect of CQ on expression of kidney damage markers. (A), representative western blots of acute kidney damage marker, NGAL and GAPDH (loading control). Black vertical lines in A show that the bands were cropped from the same blot (full blots are provided as Supplementary Figure S3). (B), graphical representation of NGAL/GAPDH normalized data. The error bar represents mean ± SEM. N = 4. (C, D), representative images of immunofluorescence of apoptotic proteins, Bax (red) and Cytochrome C (green) in cortex (C) and medulla (D) in sham, vehicle (control) and CQ. (E–H), graphs of mean fluorescence intensity of Bax in cortex (E) and medulla (F), and of Cytochrome C in cortex (G) and medulla (H). N = 4. Nuclei in all immunofluorescence microscopy images were stained with DAPI (blue). Statistical analysis- Ordinary One Way ANOVA and Fisher’s test.
FIGURE 8
FIGURE 8
Effect of CQ on mitochondrial DNA in kidneys. (A), representative in situ PCR images vehicle and CQ treated rat kidney tissues after 120 min of resuscitation. Probe represents the negative control (background fluorescence) of the in situ PCR reaction. Red fluorescence indicates amplified MT-ATP8 DNA in mitochondria. Image magnification 200x. (B), fluorescence intensity graph of MT-ATP8 DNA. N = 4. Values are expressed as mean ± SEM. Statistical analysis- Ordinary One Way ANOVA and Fisher’s test.

References

    1. Ak Ranjan M. J., Hardikar A. A. (2015). Characterizing cellular identity at one cell resolution. New York, NY: Humana Press.
    1. Bonanno F. (2011). Clinical pathology of the shock syndromes. J. Emerg. Trauma Shock 4, 233–243. 10.4103/0974-2700.82211
    1. Bonanno F. (2011). Physiopathology of shock. J. Emerg. Trauma Shock 4, 222–232. 10.4103/0974-2700.82210
    1. Briyal S., Nguyen C., Leonard M., Gulati A. (2015). Stimulation of endothelin B receptors by IRL-1620 decreases the progression of Alzheimer's disease. Neuroscience 301, 1–11. 10.1016/j.neuroscience.2015.05.044
    1. Briyal S., Ranjan A. K., Hornick M. G., Puppala A. K., Luu T., Gulati A. (2019). Anti-apoptotic activity of ETB receptor agonist, IRL-1620, protects neural cells in rats with cerebral ischemia. Sci. Rep. 9, 10439. 10.1038/s41598-019-46203-x
    1. Carrick M. M., Leonard J., Slone D. S., Mains C. W., Bar-Or D. (2016). Hypotensive resuscitation among trauma patients. Biomed. Res. Int. 2016, 8901938. 10.1155/2016/8901938
    1. de Leeuw P. W., Birkenhager W. H. (1988). Alpha-adrenoceptors and the kidney. J. Hypertens. 6, S21–S24. 10.1097/00004872-198812001-00004
    1. Faubel S. (2009). Acute kidney injury and multiple organ dysfunction syndrome. Minerva Urol. Nefrol 61, 171–188.
    1. Filho R. R., Rocha L. L., Corrêa T. D., Pessoa C. M. S., Colombo G., Assuncao M. S. C. (2016). Blood lactate levels cutoff and mortality prediction in sepsis-time for a reappraisal? A retrospective cohort study. Shock 46, 480–485. 10.1097/shk.0000000000000667
    1. Grams M. E., Rabb H. (2012). The distant organ effects of acute kidney injury. Kidney Int. 81, 942–948. 10.1038/ki.2011.241
    1. Gulati A., Arora R. C., Crayton J. (1993). Central serotonergic uptake mechanisms in hypertensive rats: effects of clonidine and centhaquin. Eur. J. Pharmacol. 231, 151–156. 10.1016/0014-2999(93)90443-l
    1. Gulati A., Choudhuri R., Gupta A., Singh S., Ali S. K. N., Sidhu G. K., et al. (2020). A multicentric, randomized, controlled phase III study of centhaquine (Lyfaquin®) as a resuscitative agent in hypovolemic shock patients. medRxiv, 2020. 10.1101/2020.07.30.20068114
    1. Gulati A., Hussain G., Srimal R. C. (1991). Effect of repeated administration of centhaquin, a centrally acting hypotensive drug, on adrenergic, cholinergic (muscarinic), dopaminergic, and serotonergic receptors in brain regions of rat. Drug Dev. Res. 23, 307–323. 10.1002/ddr.430230404
    1. Gulati A. J., Dinesh, Agrawal Nilesh., Rahate Prashant., Das Soumen., Chowdhuri Rajat., et al. (2019).Clinical phase II results OF PMZ-2010 (centhaquin) as a resuscitative agent for hypovolemic SHOCK. Crit. Care Med. 47, 1. 10.1097/01.ccm.0000550815.69306.46
    1. Gulati A., Lavhale M., Giri R., Andurkar S. V., Xanthos T. (2020). Centhaquine citrate. alpha2B-Adrenoceptor ligand, Resuscitative agent for hypovolemic shock. Drugs Fut 45, 153–163. 10.1358/dof.2020.45.3.3098155
    1. Gulati A., Lavhale M. S., Garcia D. J., Havalad S. (2012). Centhaquin improves resuscitative effect of hypertonic saline in hemorrhaged rats. J. Surg. Res. 178, 415–423. 10.1016/j.jss.2012.02.005
    1. Gulati A. (2016). Vascular endothelium and hypovolemic shock. Cvp 14, 187–195. 10.2174/1570161114666151202210221
    1. Gulati A., Zhang Z., Murphy A., Lavhale M. S. (2013). Efficacy of centhaquin as a small volume resuscitative agent in severely hemorrhaged rats. Am. J. Emerg. Med. 31, 1315–1321. 10.1016/j.ajem.2013.05.032
    1. Gutierrez G., Reines H. D., Wulf-Gutierrez M. E. (2004). Clinical review: hemorrhagic shock. Crit. Care 8, 373–381. 10.1186/cc2851
    1. Haljamäe H. (1985). Pathophysiology of shock-induced disturbances in tissue homeostasis. Acta Anaesthesiol Scand. Suppl. 29, 38–44. 10.1111/j.1399-6576.1985.tb02340.x
    1. Haljamäe H. (1993). The pathophysiology of shock. Acta Anaesthesiol Scand. Suppl. 37, 3–6. 10.1111/j.1399-6576.1991.tb05064.x
    1. Han M., Li Y., Wen D., Liu M., Ma Y., Cong B. (2018). NGAL protects against endotoxin-induced renal tubular cell damage by suppressing apoptosis. BMC Nephrol. 19, 168. 10.1186/s12882-018-0977-3
    1. Harrois A., Soyer B., Gauss T., Hamada S., Raux M., Duranteau J., et al. (2018). Prevalence and risk factors for acute kidney injury among trauma patients: a multicenter cohort study. Crit. Care 22, 344. 10.1186/s13054-018-2265-9
    1. Hekmat R., Mohebi M. (2016). Comparison of serum creatinine, cystatin C, and neutrophil gelatinase-associated lipocalin for acute kidney injury occurrence according to risk, injury, failure, loss, and end-stage criteria classification system in early after living kidney donation. Saudi J. Kidney Dis. Transpl. 27, 659–664. 10.4103/1319-2442.185220
    1. Holmquist-Mengelbier L., Fredlund E., Löfstedt T., Noguera R., Navarro S., Nilsson H., et al. (2006). Recruitment of HIF-1α and HIF-2α to common target genes is differentially regulated in neuroblastoma: HIF-2α promotes an aggressive phenotype. Cancer Cell 10, 413–423. 10.1016/j.ccr.2006.08.026
    1. Iyer N. V., Kotch L. E., Agani F., Leung S. W., Laughner E., Wenger R. H., et al. (1998). Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1alpha. Genes Develop. 12, 149–162. 10.1101/gad.12.2.149
    1. Jackson C. A., Insel P. A. (1993). Renal ?-adrenergic receptors and genetic hypertension. Pediatr. Nephrol. 7, 853–858. 10.1007/bf01213373
    1. Kimmoun A., Novy E., Auchet T., Ducrocq N., Levy B. (2015). Hemodynamic consequences of severe lactic acidosis in shock states: from bench to bedside. Crit. Care 19, 175. 10.1186/s13054-015-0896-7
    1. Ko S. F., Yip H. K., Zhen Y. Y., Lee C. C., Lee C. C., Huang S. J., et al. (2017). Severe bilateral ischemic-reperfusion renal injury: hyperacute and acute changes in apparent diffusion coefficient, T1, and T2 mapping with immunohistochemical correlations. Sci. Rep. 7, 1725. 10.1038/s41598-017-01895-x
    1. Koh M. Y., Lemos R., Jr., Liu X., Powis G. (2011). The hypoxia-associated factor switches cells from HIF-1α- to HIF-2α-Dependent signaling promoting stem cell characteristics, aggressive tumor growth and invasion. Cancer Res. 71, 4015–4027. 10.1158/0008-5472.can-10-4142
    1. Kontouli Z., Staikou C., Iacovidou N., Mamais I., Kouskouni E., Papalois A., et al. (2019). Resuscitation with centhaquin and 6% hydroxyethyl starch 130/0.4 improves survival in a swine model of hemorrhagic shock: a randomized experimental study. Eur. J. Trauma Emerg. Surg. 45, 1077–1085. 10.1007/s00068-018-0980-1
    1. Laustsen C., Lipso K., Ostergaard J. A., Nielsen P. M., Bertelsen L. B., Flyvbjerg A., et al. (2019). High intrarenal lactate production inhibits the renal pseudohypoxic response to acutely induced hypoxia in diabetes. Tomography 5, 239–247. 10.18383/j.tom.2019.00003
    1. Lavhale M. S., Havalad S., Gulati A. (2013). Resuscitative effect of centhaquin after hemorrhagic shock in rats. J. Surg. Res. 179, 115–124. 10.1016/j.jss.2012.08.042
    1. Lee D. C., Sohn H. A., Park Z.-Y., Oh S., Kang Y. K., Lee K.-m., et al. (2015). A lactate-induced response to hypoxia. Cell 161, 595–609. 10.1016/j.cell.2015.03.011
    1. Legrand M., Mik E. G., Balestra G. M., Lutter R., Pirracchio R., Payen D., et al. (2010). Fluid resuscitation does not improve renal oxygenation during hemorrhagic shock in rats. Anesthesiology 112, 119–127. 10.1097/aln.0b013e3181c4a5e2
    1. Marxsen J. H., Stengel P., Doege K., Heikkinen P., Jokilehto T., Wagner T., et al. (2004). Hypoxia-inducible factor-1 (HIF-1) promotes its degradation by induction of HIF-α-prolyl-4-hydroxylases. Biochem. J. 381, 761–767. 10.1042/bj20040620
    1. Nissanka N., Moraes C. T. (2018). Mitochondrial DNA damage and reactive oxygen species in neurodegenerative disease. FEBS Lett. 592, 728–742. 10.1002/1873-3468.12956
    1. Okusa M. D. (2010). The changing pattern of acute kidney injury: from one to multiple organ failure. Contrib. Nephrol. 165, 153–158. 10.1159/000313754
    1. World Health Organization (2019). International nonproprietary names for pharmaceutical substances (INN): proposed INN: list 121. WHO Drug Inf. 33, 233–392.
    1. Papapanagiotou P., Xanthos T., Gulati A., Chalkias A., Papalois A., Kontouli Z., et al. (2016). Centhaquin improves survival in a swine model of hemorrhagic shock. J. Surg. Res. 200, 227–235. 10.1016/j.jss.2015.06.056
    1. Ranjan A. K., Briyal S., Gulati A. (2020). Sovateltide (IRL-1620) activates neuronal differentiation and prevents mitochondrial dysfunction in adult mammalian brains following stroke. Sci. Rep. 10, 12737. 10.1038/s41598-020-69673-w
    1. Ranjan A. K., Joglekar M. V., Atre A. N., Patole M., Bhonde R. R., Hardikar A. (2012). Simultaneous imaging of microRNA or mRNA territories with protein territory in mammalian cells at single cell resolution. RNA Biol. 9, 949–953. 10.4161/rna.20484
    1. Reniguntala M. S., Lavhale M. S., Andurkar S. V., Gulati A. (2015). Synthesis and characterization of centhaquin and its citrate salt and a comparative evaluation of their cardiovascular actions. Drug Res. (Stuttg) 65, 184–191. 10.1055/s-0034-1375629
    1. Safar M. E., Struijker-Boudier H. A. (2010). Cross-talk between macro- and microcirculation. Acta Physiol. (Oxf) 198, 417–430. 10.1111/j.1748-1716.2009.02073.x
    1. Schoepflin Z. R., Silagi E. S., Shapiro I. M., Risbud M. V. (2017). PHD3 is a transcriptional coactivator of HIF‐1α in nucleus pulposus cells independent of the PKM2‐JMJD5 axis. FASEB j. 31, 3831–3847. 10.1096/fj.201601291r
    1. Semenza G. L. (2001). Hypoxia-inducible factor 1: control of oxygen homeostasis in health and disease. Pediatr. Res. 49, 614–617. 10.1203/00006450-200105000-00002
    1. Sheng Y., Zhu L. (2018). The crosstalk between autonomic nervous system and blood vessels. Int. J. Physiol. Pathophysiol Pharmacol. 10, 17–28.
    1. Srimal R. C., Gulati K., Nityanand S., Dhawan B. N. (1990). Pharmacological studies on 2-(2-(4-(3-methylphenyl)-1-piperazinyl)ethyl) quinoline (centhaquin). I. Hypotensive activity. Pharmacol. Res. 22, 319–329. 10.1016/1043-6618(90)90729-w
    1. Tisherman S. A., Schmicker R. H., Brasel K. J., Bulger E. M., Kerby J. D., Minei J. P., et al. (2015). Detailed description of all deaths in both the shock and traumatic brain injury hypertonic saline trials of the Resuscitation Outcomes Consortium. Ann. Surg. 261, 586–590. 10.1097/sla.0000000000000837
    1. Wadei H. M., Textor S. C. (2012). The role of the kidney in regulating arterial blood pressure. Nat. Rev. Nephrol. 8, 602–609. 10.1038/nrneph.2012.191
    1. Wang L., Song J., Buggs J., Wei J., Wang S., Zhang J., et al. (2017). A new mouse model of hemorrhagic shock-induced acute kidney injury. Am. J. Physiology-Renal Physiol. 312, F134–F142. 10.1152/ajprenal.00347.2016
    1. Wu L. Y., He Y. L., Zhu L. L. (2018). Possible role of PHD inhibitors as hypoxia-mimicking agents in the maintenance of neural stem cells' self-renewal properties. Front Cel Dev Biol 6, 169. 10.3389/fcell.2018.00169
    1. Xie L., Xiao K., Whalen E. J., Forrester M. T., Freeman R. S., Fong G., et al. (2009). Oxygen-regulated 2-adrenergic receptor hydroxylation by EGLN3 and ubiquitylation by pVHL. Sci. Signaling 2, ra33. 10.1126/scisignal.2000444
    1. Zakaria E. R., Garrison R. N., Spain D. A., Matheson P. J., Harris P. D., Richardson J. D. (2003). Intraperitoneal resuscitation improves intestinal blood flow following hemorrhagic shock. Ann. Surg. 237, 704–713. discussion 711-3. 10.1097/01.sla.0000064660.10461.9d
    1. Zhang J., Han J., Liu J., Liang B., Wang X., Wang C. (2017). Clinical significance of novel biomarker NGAL in early diagnosis of acute renal injury. Exp. Ther. Med. 14, 5017–5021. 10.3892/etm.2017.5150
    1. Ziello J. E., Jovin I. S., Huang Y. (2007). Hypoxia-Inducible Factor (HIF)-1 regulatory pathway and its potential for therapeutic intervention in malignancy and ischemia. Yale J. Biol. Med. 80, 51–60.

Source: PubMed

3
Sottoscrivi