Journal of Clinical Nephrology and Therapeutics

All submissions of the EM system will be redirected to Online Manuscript Submission System. Authors are requested to submit articles directly to Online Manuscript Submission System of respective journal.
Reach Us +1 (202) 780-3397

Perspective - Journal of Clinical Nephrology and Therapeutics (2023) Volume 7, Issue 6

Evaluation of novel biomarkers for early detection and monitoring of acute kidney injury in critically ill patients.

Cheng Huang*

Department of Chinese Ethnic Medicine, Guizhou Minzu University, China

*Corresponding Author:
Cheng Huang
Department of Chinese Ethnic Medicine, Guizhou Minzu University, China
Ningxia Medical University
China
E-mail:mali@mhang.cn

Received:02-Dec-2023,Manuscript No. AACNT-24-127533; Editor assigned:04-Dec-2023,PreQC No. AACNT-24-127533(PQ); Reviewed:18-Dec-2023,QC No. AACNT-24-127533; Revised:22-Dec-2023,Manuscript No. AACNT-24-127533(R); Published:30-Dec-2023,DOI: 10.35841/aacnt-7.6.179

Citation: Huang C. Evaluation of novel biomarkers for early detection and monitoring of acute kidney injury in critically ill patients. J Clin Nephrol Ther. 2023; 7(6):179

Visit for more related articles at Journal of Clinical Nephrology and Therapeutics

Introduction

In critically ill patients, acute kidney damage (AKI) is a frequent and dangerous consequence that is linked to higher rates of morbidity, death, and medical expenses. For prompt intervention and better patient outcomes, early detection and monitoring of AKI are essential. The sensitivity and specificity of conventional indicators, like urine output and serum creatinine, are not as good for early identification of AKI.[1].

Many new biomarkers have been studied in recent years in hopes of determining their usefulness for AKI monitoring and early identification. The purpose of this study is to assess the available data on new biomarkers for AKI in patients who are critically unwell. Key novel biomarkers, such as kidney injury molecule-1 (KIM-1), tissue inhibitor of neutrophil gelatinase-associated lipocalin (NGAL), interleukin-18 (IL-18), and kidney injury molecule-1 (NGAL), will be discussed along with their properties and modes of action. When kidney function rapidly declines, it can lead to compromised fluid and electrolyte balance, waste product accumulation, and disturbance of renal homeostasis. Acute kidney injury (AKI) is a common and significant consequence in critically ill patients. Because of its link to higher rates of morbidity, mortality, and medical expenses, early identification and monitoring of AKI are crucial for prompt intervention and better patient outcomes.[2].

Changes in urine output and serum creatinine levels have historically been used to diagnosis AKI. However, these markers are not ideal because of their nonspecificity and delayed reactivity, especially in patients who are critically ill and have variable kidney function. As a result, there is increasing interest in finding new biomarkers that can detect AKI earlier and more accurately, enabling prompt intervention and mitigation. Many new biomarkers have surfaced in the last few years as viable options for the early identification and tracking of AKI in individuals who are critically unwell. These biomarkers provide insights into various pathophysiological mechanisms of AKI, such as inflammation, tubular injury, and cell cycle arrest. They include neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), interleukin-18 (IL-18), tissue inhibitor of metalloproteinases-2 (TIMP-2), and insulin-like growth factor-binding protein 7 (IGFBP7) (TIMP-2*IGFBP7).[3].

Changes in urine output and serum creatinine levels have historically been used to diagnosis AKI. However, these markers are not ideal because of their nonspecificity and delayed reactivity, especially in patients who are critically ill and have variable kidney function. As a result, there is increasing interest in finding new biomarkers that can detect AKI earlier and more accurately, enabling prompt intervention and mitigation. Many new biomarkers have surfaced in the last few years as viable options for the early identification and tracking of AKI in individuals who are critically unwell. These biomarkers provide insights into various pathophysiological mechanisms of AKI, such as inflammation, tubular injury, and cell cycle arrest. They include neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), interleukin-18 (IL-18), tissue inhibitor of metalloproteinases-2 (TIMP-2), and insulin-like growth factor-binding protein 7 (IGFBP7) (TIMP-2*IGFBP7).[4].

In summary, innovative biomarkers have the potential to enhance patient outcomes and lessen the impact of this debilitating illness in the critical care setting by enabling the early detection and monitoring of AKI in critically sick patients. To fully realise the potential of these indicators in enhancing patient treatment and outcomes in AKI, more investigation and clinical validation are required.[5].

Conclusion

The present evidence on novel biomarkers for acute kidney injury (AKI) in critically ill patients has been assessed in this review, with particular attention paid to important biomarkers like tissue inhibitor of metalloproteinases-2 (TIMP-2) and insulin-like growth factor (NGAL), kidney injury molecule-1 (KIM-1), interleukin-18 (IL-18), and neutrophil gelatinase-associated lipocalin (NGAL). In comparison to conventional biomarkers, the evidence generally points to the possibility of earlier and more accurate detection of AKI using new biomarkers. They might also be useful in forecasting the course, severity, and clinical results of AKI. Still, there are a number of obstacles to overcome, such as test standardisation, clinical practice integration, and cost-effectiveness. Subsequent investigations ought to concentrate on verifying the clinical effectiveness of innovative biomarkers in various patient demographics and environments, in addition to investigating their possible contribution in steering treatment strategies and enhancing patient results in AKI. To overcome these obstacles and make it easier for novel biomarkers to be incorporated into standard clinical practice, researchers, doctors, and legislators must work together.

References

References

  1. Sigmund M, Ferstl R. Panel vector autoregression in R with the package panelvar. Q Rev Econ Finance. 2021; 80:693-720.
  2. Indexed at, Google scholar

     

  3. Yang X, Le Minh H, Cheng KT, et al. Renal compartment segmentation in DCE-MRI images. Med Image Anal. 2016;32:269-80.
  4. Indexed at, Google scholar, Cross ref

     

  5. Bhutani H, Smith V, Rahbari-Oskoui F, et al. A comparison of ultrasound and magnetic resonance imaging shows that kidney length predicts chronic kidney disease in autosomal dominant polycystic kidney disease. Kidney int. 2015;88(1):146-51.
  6. Indexed at, Google scholar, Cross ref

     

  7. Kistler AD, Poster D, Krauer F, et al. Increases in kidney volume in autosomal dominant polycystic kidney disease can be detected within 6 months. Kidney int. 2009;75(2):235-41.
  8. Indexed at, Google scholar, Cross ref

     

  9. Cardenas CE, Yang J, Anderson BM, et al. Advances in auto-segmentation. Semin radiat oncol. 2019;29(3): 185-197.
  10. Indexed at, Google scholar, Cross ref

  11. Chapman AB, Devuyst O, Eckardt KU, et al. Autosomal-dominant polycystic kidney disease (ADPKD): Executive summary from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int. 2015;88(1):17-27.
  12. Indexed at, Google scholar, Cross ref

     

  13. Kistler AD, Poster D, Krauer F, et al. Increases in kidney volume in autosomal dominant polycystic kidney disease can be detected within 6 months. Kidney int. 2009;75(2):235-41.
  14. Indexed at, Google scholar, Cross ref

     

  15. Bhutani H, Smith V, Rahbari-Oskoui F, et al. A comparison of ultrasound and magnetic resonance imaging shows that kidney length predicts chronic kidney disease in autosomal dominant polycystic kidney disease. Kidney int. 2015;88(1):146-51.
  16. Indexed at, Google scholar, Cross ref

     

  17. Momeny M, Neshat AA, Hussain MA, et al . Learning-to-augment strategy using noisy and denoised data: Improving generalizability of deep CNN for the detection of COVID-19 in X-ray images. Comput Biol Med. 2021;136:104704.
  18. Indexed at, Google scholar , Cross ref

     

  19. Sigmund M, Ferstl R. Panel vector autoregression in R with the package panelvar. Q Rev Econ Finance. 2021; 80:693-720.
  20. Indexed at, Google scholar

Get the App