GET THE APP

Prolylcarboxypeptidase in Cardiovascular Physiology and Pathophys
Journal of Clinical Toxicology

Journal of Clinical Toxicology
Open Access

ISSN: 2161-0495

+44 1478 350008

Editorial - (2012) Volume 2, Issue 8

Prolylcarboxypeptidase in Cardiovascular Physiology and Pathophysiology

Boullard N1, Madkhali H1 and Shariat-Madar Z1,2,3
1Department of Pharmacology, University of Mississippi, USA
2Research Institute of Pharmaceutical Sciences, University of Mississippi, USA
3Light Microscopy Core, University of Mississippi, USA

Keywords: Melanocortin system; Pain; Renin-Angiotensin System (RAS); Diabetes; Autophagy

Introduction

PRCP is one of the two members of the S28 exopeptidase [1] that, in addition to its effects on neuronal [2] and non-neuronal hormones [3], regulates a wide range of cell functions such as proliferation, autophagy [4], and the maintenance of vascular homeostasis (Figure 1).

clinical-toxicology-roles-mammals

Figure 1: Schematic presentation of PRCP roles in mammals This schematic presentation highlights major cell functions, which are regulated by PRCP following stimulation by hormones, neuromediators, or drugs. It appears that PRCP is highly regulated and the target of pharmaceutical intervention. AT2; angiotensin II type 2 receptor, SHP-1; sarcoma homology region 2 domain-containing phosphatase 1 (also known as tyrosin protein phosphatase type 6), LC3-2; microtubule-associated protein 1 light chain 3 type 2, BK; bradykinin, KLF2; Krueppel-like factor 2, eNOS; endothelial nitric oxide synthase, B2; bradykinin B2 receptor, MasR; Mas 1 oncogene, α-MSH; alpha melanocyte stimulating hormone, IL; interleukin, TNF-α; tumor necrosis factor alpha, IFN-γ; interferon gamma, and ICAM-1; intracellular Adhesion molecule 1.

PRCP appears to favor vasorelaxation, which contributes to normal blood flow. Impaired PRCP function is associated with cardiovascular diseases including diabetes, obesity, thrombosis and hypertension.

The biochemical aspects of PRCP were reported slightly over forty year ago [5]; however, the wide spectrum of physiological and pharmacological properties of PRCP is gradually being broadened. PRCP metabolizes Ang II to angiotensin 1-7 (Ang 1-7), stimulating the synthesis and release of the two well-known vasodilators, prostaglandin and nitric oxide (Figure 1). While much research remains to be done, studies show that PRCP metabolizes Ang III (a vasopressin peptide) to angiotensin 2-7 (Ang 2-7). Although its half-life (17 sec) is short, studies are needed to determine whether Ang 2-7 mimics Ang 1-7 as an alternative physiological adaptation. PRCP metabolizes both des-Arg [6] -bradykinin (des-Arg [6]-BK, also known as BK1-8 [7], a potent proinflammatory peptide) and alpha melanocyte stimulating hormone (α-MSH, an anti-inflammatory hormone, energy metabolism) [2,8] suggesting that PRCP inherently protects cardiovascular functioning (Figure 1).

The plasma Kallikrein-Kinin System (KKS) plays a role in the activation of blood coagulation cascade by acting as co-stimulator of tenase complex on activation of the blood clotting pathway through a unique proteolytic cascade. A major downstream effector of the KKS in activating tenase is activated factor IX (FIXa), which forms tenase complex in the presence of activated factor VIII. Prekallikrein, a key component of the KKS can be activated by a wide variety of different stimuli, including PRCP in vitro and in vivo. However, the mechanism by which PRCP-dependent PK activation occurs has not been identified. The second major effector of KKS on the surface of endothelium is bradykinin, a potent inflammatory peptide.

Activation of bradykinin 2 receptor (B2) by BK mediates release of nitric oxide (NO), prostacyclin (PGI2), and tissue plasminogen activator (tPA), which are involved in intracellular signaling protecting cells from overstretching and thrombus formation (Figure 1). PRCP induces cytoprotection through dampening reactive oxygen species (ROS) signaling (Figure 1) [9]. Although both PGI2 and NO play roles in the maintenance of vascular homeostasis, PGI2 has been found to be the major mediator of acetylcholine-induced vasodilatation in the isolated mouse heart [6]. Muscle velocity is reduced in mice with PRCP deficiency [9]. Does PRCP reduce a burden on the heart?

Peptidomic profiling of human cerebrospinal fluid and knockout mice studies have uncovered new functions for the PRCP-dependent pathway in central nervous system. Recently, YPRPIHPA (the extracellular portion of endothelin B receptor-like protein 2), a novel substrate for PRCP is found in human cerebrospinal fluid [10]. Understanding this process can provide insight into how PRCP functions in human cerebrospinal fluid. α-MSH appears to be a multifunctional neurohormone [11], which regulates a variety of physiological processes including energy balance regulation through centrally pathway and lipolysis in adipocytes [12]. PRCP metabolizes the anorexigenic α-MSH, leading to an activation of an orexigenic pathway.

Although a number of synthetic PRCP inhibitors have been developed in recent years, the endogenous inhibitor of PRCP has not yet been identified. Studies into identification of the endogenous inhibitor of PRCP may allow a better understanding of PRCP contributions to central and peripheral systems. Needless to say, a potential endogenous inhibitor of PRCP was found to be angiotensin I (Ang I) in vitro [13]. The physiological significance of this inhibition by Ang I is not fully understood. Since PRCP has a role in blood flow along with the fact that the concentration of Ang I is 45 times higher than that of Ang II in the renal vein [14], Ang I might be a major inhibitor of PRCP in kidney. Further investigations are necessary to determine if there is an association.

Emerging evidence provides support the idea that molecules, hormones and neurotransmitters known to regulated blood pressure, inflammation and pain also regulate PRCP expression and functions (Figure 1). CGP42112A is an angiotensin type 2 (AT2) receptor agonist. More recently, it has been reported that CGP42112A enhances PRCP expression and function [15]. Thus, this evidence positions PRCP downstream in AT2 signaling pathway. AT2 is upregulated in resistance arteries of hypertensive diabetic patients treated with AT1 blocker. It is reasonable to assume that PRCP expression can be influenced in diabetic patients. However, investigations are needed to validate this association.

Rutaecarpine is an alkaloid found in Evodia rutaecarpa herbs, which blocks elevated blood pressure and prevents vascular hypertrophy, also enhances expression of PRCP in the circulation and small arteries in renovascular hypertensive rats. The implications of this study for hypertensive patients are not known. However, the study suggests direct regulation of PRCP in response to the external stimuli, which will have fundamental importance for characterizing PRCP regulation under physiological condition. Glucocorticoid or its synthetic analog, dexamethasone plus [D-Ala2, N-Me-Phe4, Gly5-ol]-Enkephalin acetate increases PRCP gene in human SH-SY5Y neuroblastoma cells [16]. However, it is not known whether PRCP-dependent pathways contribute to neuronal functions and perhaps pain sensitivity.

In summary, it appears to be ubiquitous, suggesting that PRCP might have many different physiological roles. It would be an important step to definitely unravel the true extent of PRCP biological function. A protease with the ability to regulate autophagy and proliferation in addition to the inflammation, diabetes and feeding behavior proves to be extreme versatile. What are some questions that need to be answered? What other processes does PRCP affect? What controls PRCP expression? A number of intriguing clinical observations suggest that PRCP expression and function is altered in hypertension, rheumatoid arthritis, and diabetes. How exactly does altered PRCP activity contribute to disease such as rheumatoid arthritis and diabetes? Given these provocative findings, it is not clear whether aberrant PRCP activity actually affects patients’ chance of getting these diseases. However, the results of these studies suggest that the fine-tuned PRCP– dependent pathway becomes altered with the status of cell function that in part may be utilized to determine the onset and progression of cell abnormality. The emerging evidence offers a new paradigm in understanding the function of PRCP and providing implications for the pathogenesis and treatment cardiovascular disease.

Acknowledgements

This work was supported by SBAHQ-10-1-0309 to ZSM

References

  1. Kozarich JW (2010) S28 peptidases: lessons from a seemingly 'dysfunctional' family of two. BMC Biol 8: 87.
  2. Shariat-Madar B, Kolte D, Verlangieri A, Shariat-Madar Z (2010) Prolylcarboxypeptidase (PRCP) as a new target for obesity treatment. Diabetes Metab Syndr. Obes 3; 67-78.
  3. Skidgel RA, Wickstrom E, Kumamoto K, Erdös EG (1981) Rapid radioassay for prolylcarboxypeptidase (angiotensinase C). Anal Biochem 118: 113-119.
  4. Duan L, Motchoulski N, Danzer B, Davidovich I, Shariat-Madar Z, et al. (2011) Prolylcarboxypeptidase regulates proliferation, autophagy, and resistance to 4-hydroxytamoxifen-induced cytotoxicity in estrogen receptor-positive breast cancer cells. J Biol Chem 286: 2864-2876.
  5. Yang HY, Erdös EG, Chiang TS (1968) New enzymatic route for the inactivation of angiotensin. Nature 218: 1224-1226.
  6. Gwózdz P, Drelicharz L, Kozlovski VI, Chlopicki S (2007) Prostacyclin, but not nitric oxide, is the major mediator of acetylcholine-induced vasodilatation in the isolated mouse heart. Pharmacol Rep 59: 545-552.
  7. Chajkowski SM, Mallela J, Watson DE, Wang J, McCurdy CR, et al. (2011) Highly selective hydrolysis of kinins by recombinant prolylcarboxypeptidase. Biochem Biophys Res Commun 405: 338-343.
  8. Li D, Taylor AW (2008) Diminishment of alpha-MSH anti-inflammatory activity in MC1r siRNA-transfected RAW264.7 macrophages. J Leukoc Biol 84: 191-198.
  9. Adams GN, LaRusch GA, Stavrou E, Zhou Y, Nieman MT, et al. (2011) Murine prolylcarboxypeptidase depletion induces vascular dysfunction with hypertension and faster arterial thrombosis. Blood 117: 3929-3937.
  10. Zhao X, Southwick K, Cardasis HL, Du Y, Lassman ME, et al. (2010) Peptidomic profiling of human cerebrospinal fluid identifies YPRPIHPA as a novel substrate for prolylcarboxypeptidase. Proteomics 10: 2882-2886.
  11. Wu CS, Greenwood DR, Cooney JM, Jensen DJ, Tatnell MA, et al. (2006) Peripherally administered desacetyl alpha-MSH and alpha-MSH both influence postnatal rat growth and associated rat hypothalamic protein expression. Am J Physiol Endocrinol Metab 291: E1372-1380.
  12. Bradley RL, Mansfield JP, Maratos-Flier E (2005) Neuropeptides, including neuropeptide Y and melanocortins, mediate lipolysis in murine adipocytes. Obes Res 13: 653-661.
  13. Mallela J, Perkins R, Yang J, Pedigo S, Rimoldi JM, et al. (2008) The functional importance of the N-terminal region of human prolylcarboxypeptidase. Biochem Biophys Res Commun 374: 635-640.
  14. Itskovitz HD, Odya C (1971) Intrarenal formation of angiotensin I. Science 174: 58-59.
  15. Zhu L, Carretero OA, Xu J, Wang L, Harding P, et al. (2012) Angiotensin II type 2 receptor-stimulated activation of plasma prekallikrein and bradykinin release: role of SHP-1. Am J Physiol Heart Circ Physiol 302: H2553-2559.
  16. Ward GR, Franklin SO, Gerald TM, Dempsey KT, Clodfelter DE Jr, et al. (2007) Glucocorticoids plus opioids up-regulate genes that influence neuronal function. Cell Mol Neurobiol 27: 651-660.
Citation: Boullard N, Madkhali H, Shariat-Madar Z (2012) Prolylcarboxypeptidase in Cardiovascular Physiology and Pathophysiology. J Clinic Toxicol 2:e111.

Copyright: © 2012 Boullard N, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top