Perforin: An intriguing protein in allograft rejection immunology (Review)

  • Authors:
    • Ana-Maria Pașatu-Cornea
    • Elena Ciciu
    • Liliana-Ana Tuță
  • View Affiliations

  • Published online on: June 15, 2022     https://doi.org/10.3892/etm.2022.11446
  • Article Number: 519
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Chronic kidney disease (CKD) is a worldwide public health problem. The constantly increasing prevalence of CKD requires further research into new additional strategies in its management. The preferred treatment of end‑stage renal disease (ESRD) is renal transplantation. Kidney transplant patients benefit from substantial improvement in their quality and duration of life. For these to be feasible, the long‑term graft and host survival optimization of the renal transplant recipient must be ensured and chronic allograft dysfunction (CAN) must be prevented. Once an equilibrium in the allograft tolerance is established, renal transplanted patients would benefit from the withdrawal or the reduction of immunosuppression therapy. Identification of early predictive biomarkers of CAN is essential. Recent publications have revealed that in long‑term immune tolerance and graft survival several populations of immune cells are involved. Starting from the identification of perforin (PRF) in pathological renal glomeruli and following with the analysis of the molecular expression of PRF in renal biopsy samples, it appears that serum PRF is one of the potential biomarkers of graft dysfunction. Over the years, this protein has captured the attention of the medical world, conducting research that could potentially lead to the discovery of an innovative biomarker. Discovering and understanding the involvement of PRF in developing CAN may open up new therapeutic pathways that would ensure the survival of the kidney transplant. In this review the authors examined the structure, the role and the present understanding of the mechanisms by which serum PRF may be involved in chronic graft dysfunction as well as its role as an immune tolerance biomarker for chronic dysfunction of the renal graft.

1. Kidney transplantation in chronic kidney disease

Chronic kidney disease (CKD) is a worldwide public health problem. The constantly increasing prevalence of CKD requires more research into new additional strategies in its management. Impaired kidney function is accompanied by numerous complications related to water and electrolyte balance disorders and accumulation of uremic toxins which are physiologically excreted in the urine, as well as increased risk of cardiovascular events, thus affecting mortality, morbidity and the quality of life of patients with CKD (1).

The prognosis of CKD patients is dependent on the progression of renal dysfunction to its total function loss and also, on the specific complications of their chronic disease (2-4).

The optimal treatment for renal function impairment is kidney transplantation, which ensures a higher quality of life and longer survival than maintenance with dialysis, in patients with end-stage renal disease (ESRD). Over the last few years the prognosis of kidney transplanted patients has significantly improved reaching a graft survival rate of over 92% per year (5-7). Currently, the attention is directed on prolonging long-term graft survival as much as possible.

A narrative review was performed on articles published between 1980 and 2020, which were identified on PubMed, via specific mesh terms: ‘Perforin’, ‘kidney’, ‘granzyme’, ‘transplantation’, ‘graft dysfunction’. Furthermore, citation tracking of the studies retrieved was used to identify additional relevant articles. Only the articles written in English were evaluated. A total of 93 references were introduced.

2. Brief immunology of renal transplantation

Transplant rejection represents the rejection of a transplant allograft or transplanted organ. This occurs because the graft is accompanied by a series of antigens that the immune system of the recipient perceives as non-self, and consequently an immune response (host vs. graft reaction) is produced (8).

The mammalian immune system is an extremely complex system developed over millions of years as evolved immune response of vertebrates against microbial invasion and ensures the species continuity. The system could be divided into adaptive and innate immunity. Innate immunity represents a non-specific immune system, and the first line of defense that involves recruitment and participation of macrophages, neutrophils, natural killer (NK) cells, cytokines, certain cell receptors and complement components and precedes adaptive immunity functioning as secondary signals for lymphocyte activation (8).

While the inherited immunity does not involve the recognition of specific antigens, adaptive immunity involves the recognition of a wide range of molecules, the identification of different similar structures (high specificity to the pathogen agent) and the immune memory (recognition of the aggressor at the first contact and specific reaction by an accelerated and protective response) (9). The adaptive immune response is considered the most important hurdle in organ transplantation.

The main target of the immune response to the graft in organ transplantation are the major histocompatibility complex molecules (MHC) expressed on the surface of the donor cells; this feature is a form of adaptive immunity (9). The MHC is a complex of polymorphic genes encoded in a locus situated on the short arm of human chromosome 6. MHC protein products are expressed on the surfaces of various cells. In humans these are called human leukocyte antigens (HLA) and are analogous to the H-2 (in mice) and RT1 (in rats) systems (9). Graft antigens that serve as the main target of rejection are proteins encoded by the MHC genes.

Graft rejection is the result of immune mechanism activation due to antigenic differences between MHC I and II molecules of the recipient and donor, the latter acting as major antigens in the body of the recipient. These molecules present a high polymorphism, in particular those of HLA class I A and B with at least 200 and 250 alleles, respectively, which have been described in the human population while HLA-C and HLA-DP have a limited polymorphism and thus low significance (9,10).

In order to have moderate affinity for their own MHC molecules, specifically selected T cells during thymus development recognize portions of protein antigens that have been fragmented into peptides bound to MHC class I and II molecules. T-cell recognition of the antigen is the main event that initiates the effect of immune response mechanisms followed by two discrete signals. The first phase (signal 1) is the recognition of the complex formed by the MHC class II molecule and the antigenic peptide by the surface lymphocyte receptor (TCR-CD3) and Th (T helper) fixing to the antigen-presenting cell (APC). The T-cell receptor is a heterodimer consisting of an α polypeptide chain and a β polypeptide chain, which associates on the surface of the T cell, with the CD3 polypeptide complex. Signal 2 is received by the CD28 accessory molecules that bind to the B7-1 (CD80) or B7-2 (CD86) molecule on the APC surface (11,12).

Once activated T cells undergo proliferation under the influence of mitogenic growth and differentiation factors such as interleukin (IL)-2 and IL-5, which activate the target of rapamycin (TOR) paths; this process requires nucleotide synthesis. Cell proliferation and differentiation induce cytotoxicity mediated by the lymphocytes CD8+ T, activating B lymphocytes (either directly or depending on Th lymphocytes) to produce antibodies and determine macrophages to induce delayed hypersensitivity responses (11,13) (Fig. 1).

Detailed studies of these steps have led to the development of targeted immunosuppressive therapies such as IL-2 receptor blockers (basiliximab), mTOR inhibitors (sirolimus and everolimus), nucleotide synthesis inhibitors (mycophenolate) or antimetabolites (azathioprine) (14).

In 2014, a new series of small molecules with inhibiting PRF function was tested on male CD1 mice after intravenous administration; the compounds exhibited microsomal stability, which may lead to the development of a new immunosuppressive therapy (15). In contrast, the induction of PRF mRNA was partially blocked by the immunosuppressive drug cyclosporine A, and therefore this therapy has been recently avoided due to toxicity in favor of tacrolimus administration (16).

3. PRF-granzyme pathway

The main mechanism used by lymphocytes for eliminating infected or malignant cells in the host involves granule exocytosis which contains PRF and a family of serine esterases known under the name of granzymes. The fundamental basis of the apoptotic pathway PRF-granzyme, is the synergy between its components PRF and granzymes. These molecules have distinct roles. PRF pores serve as entrance gates for the protease in the targeted cell cytosol allowing granzymes to initiate various apoptotic pathways. Although granzymes can internalize independent of PRF, when seized into the endocytic vesicle lumen, they do not have access to cytosolic substrates and remain harmless. Thus, the expression of PRF is required for granule-mediated cytotoxicity, ensuring the entry of granzymes into the targeted cells, the latter to induce apoptosis (17-20) [Fig. 2, PRF-granzyme B pathway adapted from (12); T-cell immune response through the release of PRF and granzyme B, which attack target cells, inducing apoptosis.].

PRF mRNA was identified in CD8+ cells infiltrating the glomerulus of crescentic glomerulonephritis rats. In human crescentic glomerulonephritis, both CD4- and CD8-positive T lymphocytes are observed in glomeruli (21).

4. Chronic allograft dysfunction

Despite major advances in immunosuppression and transplant management, acute and chronic rejection are the main causes of kidney graft loss. It was revealed that acute rejection is the strongest predictor of subsequent chronic rejection (22).

CAN occurs due to repeated episodes of acute rejection, HLA system lack of compatibility, improper immunosuppression, ischemic injury (vascular occlusion caused by arterial immune-mediated thickening and dysfunction) with secondary fibrosis and late recovery of renal function (23).

Early allograft lesions by the PRF-granzyme pathway could initiate the development of CAN in renal allografts (24). CD4+ Th lymphocytes reactive to graft alloantigens produce cytokines which induce endothelial and smooth muscle cell proliferation and cause occlusion of the vascular lumen. It appears that cytotoxic T lymphocytes infiltrated in the renal graft induce allogeneic tubular epithelial cell death, via the native PRF pathway (25,26).

The diagnosis of chronic rejection is not always made in a timely manner, required for the complete recuperation of renal function. This problem occurs since kidney damage is detected mainly as an increase in serum creatinine and the appearance of proteinuria. However, an increase in serum creatinine is a late sign of kidney damage, as the compensatory mechanisms in the kidneys can maintain the glomerular filtration rate (GFR) despite progressive structural damage. Although the long-term survival of allografts is improving, late graft loss from CAN remains a clinically significant problem and is the second most common cause of late renal allograft loss, after death (27). Thus, the identification of early markers associated with, or predicting CAN would be clinically useful.

Recurrent or chronic inflammatory processes are common in people with CKD and particularly in those with ESRD. This is due to numerous underlying factors including the uremic environment, high levels of circulating proinflammatory cytokines, oxidative stress, carbonyl stress, waste of protein energy (PEW), and increased incidence of infections (especially dialysis access) to mention a few (28).

The acute-phase response is a major pathophysiological phenomenon that accompanies inflammation. With this reaction, normal homeostatic mechanisms are replaced by new established factors that are likely to contribute to defensive or adaptive capabilities (29,30).

In patients returning to a dialysis program after acute or chronic rejection of the renal transplant but without kidney transplant graftectomy performed, a chronic inflammatory state was observed, that was reduced by the removal of the non-functional graft (explant) (31).

5. Predictive markers of chronic allograft dysfunction

To date, few studies have been performed on PRF in the renal area. In an experimental study, using Wistar-Kyoto (WKY) rats with antiglomerular basement membrane (GBM) crescentic glomerulonephritis (GN), PRF protein and mRNA expression of PRF were demonstrated in glomeruli by immunohistochemistry and in situ hybridization. WKY rats treated with anti-PRF antibodies revealed significantly reduced amounts of proteinuria and frequency of crescentic glomeruli (21).

There is strong scientific evidence that immunological non-invasive monitoring could be useful in the first 6 months after kidney transplantation in particular regarding prediction of acute rejection episodes (32). It is less clear whether CAN is also associated with consistent changes of peripheral blood or the urinary cells. Several histological studies have demonstrated enhanced expression of granzymes and PRF in numerous types of transplanted grafts and their correlation with acute rejection episodes (33-40). It appears that the urinary mRNA levels of three markers including PRF, granzyme B and FAS ligand appear to be correlated with acute rejection and the increase of serum creatinine (41,42). The molecular analysis of the expression of these three molecules in renal biopsy samples revealed that only the expression of PRF and FAS ligand were correlated with the acute rejection while the expression of PRF and granzyme B could intensify at a longer time after transplantation, possibly associated with chronic dysfunction (43). In 1997, a concurrent RT-qPCR assessment of PRF, granzyme B and Fas ligand revealed a correlation with acute rejection even in cases of mild infiltration, with 100% sensitivity and specificity. The combined analysis of the expression of Fas ligand, PRF and granzyme B genes by quantitative RT-PCR provided a reliable tool for the diagnosis and management of acute renal rejection and antirejection therapy leading to a rapid decrease in the expression of these genes (44). Li et al confirmed that the mRNA levels of PRF and granzyme B were increased in urine samples from patients with acute rejection (42). To date, it has been demonstrated that urinary mRNA levels of AGT, EGFR and TGF-β1 may be reliable prediction markers of CAN (32,45-47).

The genetic expression of serum PRF has been revealed to be more correlated with acute rejection in renal transplanted patients in comparison with granzyme B and Fas ligand thus supporting its use as a marker of acute rejection (48). The PRF-granzyme and the Fas ligand are two major pathways by which cytotoxic T lymphocytes induce apoptosis in target cells (49). The expression of the message into the graft for these immune-activating genes has been revealed to be markedly correlated with graft rejection. In a retrospective pilot study, 140 fine-needle aspiration biopsy samples from 50 human renal allografts were labeled using alkaline phosphatase/alkaline anti-phosphatase immunocytochemistry incorporating monoclonal antibodies to PRF, granzyme B, and Fas ligand. Positive labeling levels for these markers were compared with the initial clinical diagnosis of rejection. Only when all three antibodies yielded positive labeling, was the association with the clinical rejection status superior to conventional morphological cytology (50).

A recent study on humanized mice, revealed that granzyme B expression was significantly increased in CD8+ T cells in patients with graft rejection, while surviving graft patients expressed less granzyme B, as they had an increased level of HLA-G dimer which inhibited cytotoxicity of CD8+ T cells (51). The synergy between PRF and granzymes is already established. Therefore, a high level of granzymes implies a high level of PRF.

Recently, new categories of drugs, such as inhibitors of PRF, have aroused the interest of researchers. Tampio et al demonstrated that using L-type amino acid transporter 1 (LAT1)-utilizing prodrugs of PRF inhibitors for improved administration of brain drug delivery, led to improved pharmacological effects, decreased production of cellular apoptosis mediators, decreased overall oxidative stress and inflammation in the brain, and from the periphery, increasing cell survival (52).

6. PRF ‘roots’

In the evolution of PRF there were complex models of events of birth and death including duplication/pseudogenization to mammals, multiple amplifications and losses in reptiles and fish as well and a case of partial duplication with a new beginning codon to fish. Approximately 500 million years ago, the primordial PRF gene evolved, around the same time as T-cell receptor antigen recognition based on the major histocompatibility complex. As it is absent from primitive chordates and invertebrates, cytotoxic cells from these lineages must have a different cytotoxic effector molecule or mechanism. Orthologs and homologues of human PRF have been identified in almost all species. Research has shown that in species prior to Gnathostomata (Euteleostomi) the PRF gene did not exist which suggests that cytotoxic cells of prior species have another mechanism or different means for killing targeted cells. In addition, there is evidence that PRF originated from the duplication of the ancient gene MPEG1 and shares a common ancestor with functionally related complement proteins (53,54).

7. PRF: Structure and genetics

PRF is a 67-kDa pore-forming protein, stored and released from the secretory granules (SG) of the cytotoxic lymphocytes which leads to osmotic lysis of the membrane of target cells and subsequently allows proapoptotic granzymes (serine proteases which split the peptide connections of proteins) with broad specificity to enter the targeted cells and activate the cell death program. PRF expression is increased in the activated CD8+ cells, αγ T cells, in subpopulations of activated CD4+ T cells, and NK cells (but with a high and stable incorporation in NK cells) (17,19,55-58). In addition, PRF expression may be stimulated in some activated CD4+ cells (59,60).

In mammals, PRF is encoded by the PRF1 gene expressed in cytotoxic lymphocytes and regulatory T cells. PRF1 transcription is the main mechanism that determines PRF expression in cytotoxic T lymphocytes and NK cells. While PRF is uniformly expressed by mature NK cells as a result of spontaneous stimulation of constitutive gene transcription, its expression in peripheral T cells requires gene activation (17,58,61-64).

Locus control position is essential for PRF1-specific activity (NK and cytotoxic cell activation). A heterochromatin-dependent regulation could allow certain exogenous stimuli and certain endogenous controlling of the transcription factors to induce PRF1 transcription in other types of cells (59). In 2006, a study conducted by Pipkin and Lichtenheld identified the locus control region for perforin of 150 Kb of cis action sequences which leads to the physiological PRF1 transcription, comprising 16 hypersensitive DNase I (DHS) sites, four of them necessary for PRF expression (17,65). PRF was identified for the first time in 1983 and it was cloned from an expression library by a cross reaction of the C9 antibody (59,66-71). Fine-resolution comparisons by direct sequence comparisons have revealed a similarity between the two proteins (C9 and PRF), that contain in the middle part of their sequences a short region called membrane-attack complex/PRF (MACPF) (59,72,73). Both proteins polymerize in tubular complexes able to determine lysis of the membrane insertion acting as large and voltage-independent transmembrane channels. Initial studies have revealed that while C9 polymerizes in physiological conditions requiring the assembly of complex C5b-8 into the receptor, the functional activity of PRF in the phospholipid membrane is calcium-dependent (67,74,75). After exocytosis, granules from the killer cells releasing PRF and granzymes are exposed to immune synapses rich in calcium and neutral pH (59,64). The PRF monomers bind to calcium by its C2 domain acquiring the capacity of bounding lipids to the targeted cell membrane and then to merge in transmembrane pores of up to 100 Å, which allows granzymes access to the protein substrates involved in apoptosis (59,76-78).

8. Perforinopathies

It is recognized that the residual function of the PRF-dependent cytotoxic cells causes transplant rejection of allogenic stem cells, allografts and solid organs. At the opposite pole defects of the cytotoxic path and PRF deficiency (failure to deliver PRF) lead to disorders called perforinopathies including familial haemophagocytic lymphohistiocytosis (FHL), viral infections and the predisposition to develop haemato-oncological diseases (12,79-83). Voskoboinik et al have proposed the term of perforinopathies in order to define a spectrum of immune-mediated disease responses associated with monoallelic mutations in genes related to FHL (84).

The complete absence of the PRF function results into FHL, an immunoregulatory disease that appears in childhood and is characterized by uncontrolled activation of CPA and CD8+ T lymphocytes with secondary accumulation of T cells. Recently it was discovered that the partial loss of PRF function is strongly associated with FHL and a series of hematological disorders that appear later in childhood or in adolescence. In addition, PRF functionality is essential for cytotoxic lymphocytes in humans since harmful mutations in PRF1 leads to FHL2 representing 30-60% of FHL cases (59,79,85).

PRF and CD107a tests are more sensitive and have a similar specificity compared with NK cytotoxicity test and would be able to enhance FHL screening (86).

Relative recent studies have revealed that UVB and UVA radiations induce accumulation of granzyme B in human keratinocytes. In addition, granzyme B secondary to UVB radiation mediates cytotoxicity of keratinocytes, while in UVA irradiation it increases the ability of the keratinocyte to degrade matrix extracellular components; these observations could be the basis of photoaging and photocarcinogenicity domain (87,88).

While cancer therapy has begun to use ‘suicide genes’ to induce cell apoptosis, the role of vaccination with apoptotic cells, either immune stimulatory or immune suppressive is still debated. Recently a new technology called cytolytic DNA technology has been developed, using a vaccine which encodes truncated PRF incorporated in a bicistronic DNA vector that activates dendritic cells thus stimulating the CD8+ T-cell response against HIV and HCV reducing the viral loads similar to traditional vaccines (89-92).

PRF has gained the attention of cardiology researchers, proving to date, that patients with left ventricular dysfunction have PRF-positive infiltration of heart cells and that PRF could be an adverse predictor of long-term mortality in patients with inflammatory cardiomyopathy (93).

9. Conclusions

PRF is a pore-forming protein vital for cytotoxic effector function and has an indispensable role in granzyme-mediated apoptosis. It is responsible for endothelial damage and plays a role in the pathogenesis of numerous inflammatory diseases and targets cell apoptosis.

Over the last few years, the study of serum PRF and its role in inflammatory and neoplastic diseases has captured the attention of the medical world. To date, few studies have reported the correlation between serum or urinary PRF, granzyme and ligand FAS with acute transplant rejection. Further studies are required to clarify the role of PRF as a potential early biomarker with a predictive role in chronic allograft rejection.

Advances in immunosuppressive therapy, in order to maintain kidney transplantation and avoid rejection, have led to decreased rejection rates. However, these agents are not deprived of side effects.

The residual function of PRF-dependent cytotoxic cells causes transplant rejection of allogenic stem cells, the allograft and solid organs. A deep understanding of the role of PRF in inducing allograft rejection is necessary for the development of new targeted post-transplant therapies. Highly specific inhibitors of PRF function, are thus of interest as selective immunosuppressive drugs.

The practical use of PRF expression has already been demonstrated in various medical fields. Demonstrating the utility of PRF as a predictive marker of CAN, such as with PRF inhibitors, would have treatment implications that could mark the beginning of a new era in immunosuppressive therapy.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the project ANTREPRENORDOC, in the framework of Human Resources Development Operational Program 2014-2020, financed from the European Social Fund under the contract number 36355/23.05.2019 HRD OP/380/6/13-SMIS Code: 123847.

Availability of data and materials

Not applicable.

Authors' contributions

AMPC conceived, wrote and edited the manuscript. EC reviewed the manuscript for important intellectual content. LAT revised the study before finally approving it for publication. AMPC, EC and LAT confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Covic A, Mircescu G, Gluhovschi G and Schiller A: Ghiduri de practica medicala. Boala cronica de rinichi. 1-109. 2007. Bucuresti AG de practica medicala. SR de N: No Title.

2 

Evans RW, Manninen DL, Garrison LP Jr, Hart LG, Blagg CR, Gutman RA, Hull AR and Lowrie EG: The quality of life of patients with end-stage renal disease. N Engl J Med. 312:553–559. 1985.PubMed/NCBI View Article : Google Scholar

3 

Laupacis A, Keown P, Pus N, Krueger H, Ferguson B, Wong C and Muirhead N: A study of the quality of life and cost-utility of renal transplantation. Kidney Int. 50:235–242. 1996.PubMed/NCBI View Article : Google Scholar

4 

Simmons RG and Abress L: Quality-of-life issues for end-stage renal disease patients. Am J Kidney Dis. 15:201–208. 1990.PubMed/NCBI View Article : Google Scholar

5 

Ojo AO, Hanson JA, Wolfe RA, Leichtman AB, Agodoa LY and Port FK: Long-term survival in renal transplant recipients with graft function. Kidney Int. 57:307–313. 2000.PubMed/NCBI View Article : Google Scholar

6 

Organ procurement and transplantation network and scientific registry of transplant recipients 2010 data report. Am J Transplant. 12 (Suppl 1):S1–S156. 2012.PubMed/NCBI View Article : Google Scholar

7 

Foroutan F, Friesen EL, Clark KE, Motaghi S, Zyla R, Lee Y, Kamran R, Ali E, De Snoo M, Orchanian-Cheff A, et al: Risk factors for 1-year graft loss after kidney transplantation systematic review and meta-analysis. Clin J Am Soc Nephrol. 14:1642–1650. 2019.PubMed/NCBI View Article : Google Scholar

8 

Wyburn KR, Jose MD, Wu H, Atkins RC and Chadban SJ: The role of macrophages in allograft rejection. Transplantation. 80:1641–1647. 2005.PubMed/NCBI View Article : Google Scholar

9 

Williams TM: Human leukocyte antigen gene polymorphism and the histocompatibility laboratory. J Mol Diagnostics. 3:98–104. 2001.PubMed/NCBI View Article : Google Scholar

10 

Horton R, Wilming L, Rand V, Lovering RC, Bruford EA, Khodiyar VK, Lush MJ, Povey S, Talbot CC Jr, Wright MW, et al: Gene map of the extended human MHC. Nat Rev Genet. 5:889–899. 2004.PubMed/NCBI View Article : Google Scholar

11 

Sayegh MH and Turka LA: The role of T-cell costimulatory activation pathways in transplant rejection. N Engl J Med. 338:1813–1821. 1998.PubMed/NCBI View Article : Google Scholar

12 

Osińska I, Popko K and Demkow U: Perforin: An important player in immune response. Cent Eur J Immunol. 39:109–115. 2014.PubMed/NCBI View Article : Google Scholar

13 

Vella J: Transplantation immunobiology. UpToDate, 2021.

14 

Mukherjee S and Mukherjee U: A comprehensive review of immunosuppression used for liver transplantation. J Transplant. 2009(701464)2009.PubMed/NCBI View Article : Google Scholar

15 

Bull MR, Spicer JA, Huttunen KM, Denny WA, Ciccone A, Browne KA, Trapani JA and Helsby NA: The preclinical pharmacokinetic disposition of a series of perforin-inhibitors as potential immunosuppressive agents. Eur J Drug Metab Pharmacokinet. 40:417–425. 2015.PubMed/NCBI View Article : Google Scholar

16 

Lu P, Garcia-Sanz JA, Lichtenheld MG and Podack ER: Perforin expression in human peripheral blood mononuclear cells: Definition of an IL-2-independent pathway of perforin induction in CD8+ T cells. J Immunol. 148:3354–3360. 1992.PubMed/NCBI

17 

Pipkin ME, Ljutic B, Cruz-Guilloty F, Nouzova M, Rao A, Zúñiga-Pflücker JC and Lichtenheld MG: Chromosome transfer activates and delineates a locus control region for perforin. Immunity. 26:29–41. 2007.PubMed/NCBI View Article : Google Scholar

18 

Keefe D, Shi L, Feske S, Massol R, Navarro F, Kirchhausen T and Lieberman J: Perforin triggers a plasma membrane-repair response that facilitates CTL induction of apoptosis. Immunity. 23:249–262. 2005.PubMed/NCBI View Article : Google Scholar

19 

Kägi D, Ledermann B, Bürki K, Zinkernagel RM and Hengartner H: Molecular mechanisms of lymphocyte-mediated cytotoxicity and their role in immunological protection and pathogenesis in vivo. Annu Rev Immunol. 14:207–232. 1996.PubMed/NCBI View Article : Google Scholar

20 

Henkart PA: Mechanism of lymphocyte-mediated cytotoxicity. Annu Rev Immunol. 3:31–58. 1985.PubMed/NCBI View Article : Google Scholar

21 

Fujinaka H, Yamamoto T, Feng L, Nameta M, Garcia G, Chen S, El-shemi AA, Ohshiro K, Katsuyama K, Yoshida Y, et al: Anti-perforin antibody treatment ameliorates experimental crescentic glomerulonephritis in WKY rats. Kidney Int. 72:823–830. 2007.PubMed/NCBI View Article : Google Scholar

22 

Cecka JM, Cho YW and Terasaki PI: Analyses of the UNOS scientific renal transplant registry at three years-early events affecting transplant success. Transplantation. 53:59–63. 1992.PubMed/NCBI View Article : Google Scholar

23 

Mitchell RN and Libby P: Vascular remodeling in transplant vasculopathy. Circ Res. 100:967–978. 2007.PubMed/NCBI View Article : Google Scholar

24 

Almond PS, Matas A, Gillingham K, Dunn DL, Payne WD, Gores P, Gruessner R and Najarian JS: Risk factors for chronic rejection in renal allograft recipients. Transplantation. 55:752–756; discussion 756-7. 1993.PubMed/NCBI View Article : Google Scholar

25 

Miltenburg AM, Meijer-Paape ME, Daha MR, van Bockel JH, Weening JJ, van Es LA and van*der*Woude FJ: Donor-specific lysis of human kidney proximal tubular epithelial cells by renal allograft-infiltrating lymphocytes. Transplantation. 48:296–302. 1989.PubMed/NCBI View Article : Google Scholar

26 

Wever PC, Boonstra JG, Laterveer JC, Hack CE, van Der Woude FJ, Daha MR and ten*Berge IJ: Mechanisms of lymphocyte-mediated cytotoxicity in acute renal allograft reaction. Transplantation. 66:259–264. 1998.PubMed/NCBI View Article : Google Scholar

27 

Cecka JM: The UNOS scientific renal transplant registry-2000. Clin Transpl. 1-18:2000.PubMed/NCBI

28 

Owen WF and Lowrie EG: C-reactive protein as an outcome predictor for maintenance hemodialysis patients. Kidney Int. 54:627–636. 1998.PubMed/NCBI View Article : Google Scholar

29 

Guessous I, Ponte B, Marques-Vidal P, Paccaud F, Gaspoz JM, Burnier M, Waeber G, Vollenweider P and Bochud M: Clinical and biological determinants of kidney outcomes in a population-based cohort study. Kidney Blood Press Res. 39:74–85. 2014.PubMed/NCBI View Article : Google Scholar

30 

Csaba P, Kovesdy*Kopple JD and Kalantar-Zadeh K: Inflammation in renal insufficiency. UpToDate, 2011.

31 

López-Gómez JM, Pérez-Flores I, Jofré R, Carretero D, Rodríguez-Benitez P, Villaverde M, Pérez-García R, Nassar GM, Niembro E and Ayus JC: Presence of a failed kidney transplant in patients who are on hemodialysis is associated with chronic inflammatory state and erythropoietin resistance. J Am Soc Nephrol. 15:2494–2501. 2004.PubMed/NCBI View Article : Google Scholar

32 

Mas VR, Mas LA, Archer KJ, Yanek K, King AL, Gibney EM, Cotterell A, Fisher RA, Posner M and Maluf DG: Evaluation of gene panel mrnas in urine samples of kidney transplant recipients as a non-invasive tool of graft function. Mol Med. 13:315–324. 2007.PubMed/NCBI View Article : Google Scholar

33 

Choy JC: Granzymes and perforin in solid organ transplant rejection. Cell Death Differ. 17:567–576. 2010.PubMed/NCBI View Article : Google Scholar

34 

Hameed A, Truong LD, Price V, Kruhenbuhl O and Tschopp J: Immunohistochemical localization of granzyme B antigen in cytotoxic cells in human tissues. Am J Pathol. 138:1069–1075. 1991.PubMed/NCBI

35 

Griffiths GM, Namikawa R, Mueller C, Liu CC, Young JD, Billingham M and Weissman I: Granzyme A and perforin as markers for rejection in cardiac transplantation. Eur J Immunol. 21:687–692. 1991.PubMed/NCBI View Article : Google Scholar

36 

Ciément MV, Haddad P, Soulié A, Benvenuti C, Lichtenheld MG, Podack ER, Sigaux N and Sasportes M: Perform and granzyme B as markers for acute rejection in heart transplantation. Int Immunol. 3:1175–1181. 1991.PubMed/NCBI View Article : Google Scholar

37 

Lipman ML, Stevens AC and Strom TB: Heightened intragraft CTL gene expression in acutely rejecting renal allografts. J Immunol. 152:5120–5127. 1994.PubMed/NCBI

38 

Kummer JA, Wever PC, Kamp AM, ten Berge IJ, Hack CE and Weening JJ: Expression of granzyme A and B proteins by cytotoxic lymphocytes involved in acute renal allograft rejection. Kidney Int. 47:70–77. 1995.PubMed/NCBI View Article : Google Scholar

39 

Krams SM, Villanueva JC, Quinn MB and Martinez OM: Expression of the cytotoxic T cell mediator granzyme B during liver allograft rejection. Transpl Immunol. 3:162–166. 1995.PubMed/NCBI View Article : Google Scholar

40 

Legros-Maïda S, Soulié A, Benvenuti C, Wargnier A, Vallée N, Berthou C, Guillet J, Sasportes M and Sigaux N: Granzyme B and perforin can be used as predictive markers of acute rejection in heart transplantation. Eur J Immunol. 24:229–233. 1994.PubMed/NCBI View Article : Google Scholar

41 

Yannaraki M, Rebibou JM, Ducloux D, Saas P, Duperrier A, Felix S, Rifle G, Chalopin JM, Hervé P, Tiberghien P and Ferrand C: Urinary cytotoxic molecular markers for a noninvasive diagnosis in acute renal transplant rejection. Transpl Int. 19:759–768. 2006.PubMed/NCBI View Article : Google Scholar

42 

Li B, Hartono C, Ding R, Sharma VK, Ramaswamy R, Qian B, Serur D, Mouradian J, Schwartz JE and Suthanthiran M: Noninvasive diagnosis of renal-allograft rejection by measurement of messenger RNA for perforin and granzyme B in urine. N Engl J Med. 344:947–954. 2001.PubMed/NCBI View Article : Google Scholar

43 

Graziotto R, Del Prete D, Rigotti P, Anglani F, Baldan N, Furian L, Valente M, Antonello A, Marchini F, D'Angelo A and Gambaro G: Perforin, Granzyme B, and Fas ligand for molecular diagnosis of acute renal-allograft rejection: Analyses on serial biopsies suggest methodological issues. Transplantation. 81:1125–1132. 2006.PubMed/NCBI View Article : Google Scholar

44 

Strehlau J, Pavlakis M, Lipman M, Shapiro M, Vasconcellos L, Harmon W and Strom TB: Quantitative detection of immune activation transcripts as a diagnostic tool in kidney transplantation. Proc Natl Acad Sci USA. 94:695–700. 1997.PubMed/NCBI View Article : Google Scholar

45 

Campistol JM, Iñigo P, Larios S, Bescos M and Oppenheimer F: Role of transforming growth factor-beta1 in the progression of chronic allograft nephropathy. Nephrol Dial Transplant. 16 (Suppl 1):S114–S116. 2001.PubMed/NCBI View Article : Google Scholar

46 

Nocera A, Tagliamacco A, De Palma R, Del Galdo F, Ferrante A, Fontana I, Barocci S, Ginevri F, Rolla D, Ravetti JL and Valente U: Cytokine mRNA expression in chronically rejected human renal allografts. Clin Transplant. 18:564–570. 2004.PubMed/NCBI View Article : Google Scholar

47 

Pribylova-Hribova P, Kotsch K, Lodererova A, Viklicky O, Vitko S, Volk HD and Lacha J: TGF-beta1 mRNA upregulation influences chronic renal allograft dysfunction. Kidney Int. 69:1872–1879. 2006.PubMed/NCBI View Article : Google Scholar

48 

Shin GT, Kim SJ, Lee TS, Oh CK and Kim H: Gene expression of perforin by peripheral blood lymphocytes as a marker of acute rejection. Nephron Clin Pract. 100:c63–c70. 2005.PubMed/NCBI View Article : Google Scholar

49 

Trapani JA and Smyth MJ: Functional significance of the perforin/granzyme cell death pathway. Nat Rev Immunol. 2:735–747. 2002.PubMed/NCBI View Article : Google Scholar

50 

Pascoe MD, Marshall SE, Welsh KI, Fulton LM and Hughes DA: Increased accuracy of renal allograft rejection diagnosis using combined perforin, granzyme B, and Fas ligand fine-needle aspiration immunocytology. Transplantation. 69:2547–2553. 2000.PubMed/NCBI View Article : Google Scholar

51 

Ajith A, Portik-Dobos V, Nguyen-Lefebvre AT, Callaway C, Horuzsko DD, Kapoor R, Zayas C, Maenaka K, Mulloy LL and Horuzsko A: HLA-G dimer targets Granzyme B pathway to prolong human renal allograft survival. FASEB J. 33:5220–5236. 2019.PubMed/NCBI View Article : Google Scholar

52 

Tampio J, Huttunen J, Montaser A and Huttunen KM: Targeting of perforin inhibitor into the brain parenchyma via a prodrug approach can decrease oxidative stress and neuroinflammation and improve cell survival. Mol Neurobiol. 57:4563–4577. 2020.PubMed/NCBI View Article : Google Scholar

53 

D'Angelo ME, Dunstone MA, Whisstock JC, Trapani JA and Bird PI: Perforin evolved from a gene duplication of MPEG1, followed by a complex pattern of gene gain and loss within Euteleostomi. BMC Evol Biol. 12(59)2012.PubMed/NCBI View Article : Google Scholar

54 

Araujo-Voces M and Quesada V: Frequent birth-and-death events throughout perforin-1 evolution. BMC Evol Biol. 20(135)2020.PubMed/NCBI View Article : Google Scholar

55 

De Rosa SC, Andrus JP, Perfetto SP, Mantovani JJ, Herzenberg LA, Herzenberg LA and Roederer M: Ontogeny of gamma delta T cells in humans. J Immunol. 172:1637–1645. 2004.PubMed/NCBI View Article : Google Scholar

56 

Grossman WJ, Verbsky JW, Barchet W, Colonna M, Atkinson JP and Ley TJ: Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity. 21:589–601. 2004.PubMed/NCBI View Article : Google Scholar

57 

Gumperz JE, Miyake S, Yamamura T and Brenner MB: Functionally distinct subsets of CD1d-restricted natural killer T cells revealed by CD1d tetramer staining. J Exp Med. 195:625–636. 2002.PubMed/NCBI View Article : Google Scholar

58 

Nakata M, Kawasaki A, Azuma M, Tsuji K, Matsuda H, Shinkai Y, Yagita H and Okumura K: Expression of perforin and cytolytic potential of human peripheral blood lymphocyte subpopulations. Int Immunol. 4:1049–1054. 1992.PubMed/NCBI View Article : Google Scholar

59 

Brennan AJ, Chia J, Trapani JA and Voskoboinik I: Perforin deficiency and susceptibility to cancer. Cell Death Differ. 17:607–615. 2010.PubMed/NCBI View Article : Google Scholar

60 

Voskoboinik I, Smyth MJ and Trapani JA: Perforin-mediated target-cell death and immune homeostasis. Nat Rev Immunol. 6:940–952. 2006.PubMed/NCBI View Article : Google Scholar

61 

Salcedo TW, Azzoni L, Wolf SF and Perussia B: Modulation of perforin and granzyme messenger RNA expression in human natural killer cells. J Immunol. 151:2511–2520. 1993.PubMed/NCBI

62 

Zhang J, Scordi I, Smyth MJ and Lichtenheld MG: Interleukin 2 receptor signaling regulates the perforin gene through signal transducer and activator of transcription (Stat)5 activation of two enhancers. J Exp Med. 190:1297–1308. 1999.PubMed/NCBI View Article : Google Scholar

63 

García-Sanz JA and Podack ER: Regulation of perforin gene expression in a T cell hybrid with inducible cytolytic activity. Eur J Immunol. 23:1877–1883. 1993.PubMed/NCBI View Article : Google Scholar

64 

Uellner R, Zvelebil MJ, Hopkins J, Jones J, MacDougall LK, Morgan BP, Podack E, Waterfield MD and Griffiths GM: Perforin is activated by a proteolytic cleavage during biosynthesis which reveals a phospholipid-binding C2 domain. EMBO J. 16:7287–7296. 1997.PubMed/NCBI View Article : Google Scholar

65 

Pipkin ME and Lichtenheld MG: A reliable method to display authentic DNase I hypersensitive sites at long-ranges in single-copy genes from large genomes. Nucleic Acids Res. 34(e34)2006.PubMed/NCBI View Article : Google Scholar

66 

Podack ER and Dennert G: Assembly of two types of tubules with putative cytolytic function by cloned natural killer cells. Nature. 302:442–445. 1983.PubMed/NCBI View Article : Google Scholar

67 

Podack ER, Young JD and Cohn ZA: Isolation and biochemical and functional characterization of perforin 1 from cytolytic T-cell granules. Proc Natl Acad Sci USA. 82:8629–8633. 1985.PubMed/NCBI View Article : Google Scholar

68 

Young JD, Cohn ZA and Podack ER: The ninth component of complement and the pore-forming protein (perforin 1) from cytotoxic T cells: Structural, immunological, and functional similarities. Science. 233:184–190. 1986.PubMed/NCBI View Article : Google Scholar

69 

Young JD, Hengartner H, Podack ER and Cohn ZA: Purification and characterization of a cytolytic pore-forming protein from granules of cloned lymphocytes with natural killer activity. Cell. 44:849–859. 1986.PubMed/NCBI View Article : Google Scholar

70 

Lowrey DM, Aebischer T, Olsen K, Lichtenheld M, Rupp F, Hengartner H and Podack ER: Cloning, analysis, and expression of murine perforin 1 cDNA, a component of cytolytic T-cell granules with homology to complement component C9. Proc Natl Acad Sci USA. 86:247–251. 1989.PubMed/NCBI View Article : Google Scholar

71 

Shinkai Y, Takio K and Okumura K: Homology of perforin to the ninth component of complement (C9). Nature. 334:525–527. 1988.PubMed/NCBI View Article : Google Scholar

72 

Tschopp J, Masson D and Stanley KK: Structural/functional similarity between proteins involved in complement- and cytotoxic T-lymphocyte-mediated cytolysis. Nature. 322:831–834. 1986.PubMed/NCBI View Article : Google Scholar

73 

Lichtenheld MG, Olsen KJ, Lu P, Lowrey DM, Hameed A, Hengartner H and Podack ER: Structure and function of human perforin. Nature. 335:448–451. 1988.PubMed/NCBI View Article : Google Scholar

74 

Blumenthal R, Millard PJ, Henkart MP, Reynolds CW and Henkart PA: Liposomes as targets for granule cytolysin from cytotoxic large granular lymphocyte tumors. Proc Natl Acad Sci USA. 81:5551–5555. 1984.PubMed/NCBI View Article : Google Scholar

75 

Henkart PA, Yue CC, Yang J and Rosenberg SA: Cytolytic and biochemical properties of cytoplasmic granules of murine lymphokine-activated killer cells. J Immunol. 137:2611–2617. 1986.PubMed/NCBI

76 

Müllbacher A, Waring P, Tha Hla R, Tran T, Chin S, Stehle T, Museteanu C and Simon MM: Granzymes are the essential downstream effector molecules for the control of primary virus infections by cytolytic leukocytes. Proc Natl Acad Sci USA. 96:13950–13955. 1999.PubMed/NCBI View Article : Google Scholar

77 

Nakajima H, Park HL and Henkart PA: Synergistic roles of granzymes A and B in mediating target cell death by rat basophilic leukemia mast cell tumors also expressing cytolysin/perforin. J Exp Med. 181:1037–1046. 1995.PubMed/NCBI View Article : Google Scholar

78 

Shi L, Mai S, Israels S, Browne K, Trapani JA and Greenberg AH: Granzyme B (GraB) autonomously crosses the cell membrane and perforin initiates apoptosis and GraB nuclear localization. J Exp Med. 185:855–866. 1997.PubMed/NCBI View Article : Google Scholar

79 

Spicer BA, Conroy PJ, Law RHP, Voskoboinik I and Whisstock JC: Perforin-A key (shaped) weapon in the immunological arsenal. Semin Cell Dev Biol. 72:117–123. 2017.PubMed/NCBI View Article : Google Scholar

80 

Voskoboinik I, Whisstock JC and Trapani JA: Perforin and granzymes: Function, dysfunction and human pathology. Nat Rev Immunol. 15:388–400. 2015.PubMed/NCBI View Article : Google Scholar

81 

Jenkins MR, Rudd-Schmidt JA, Lopez JA, Ramsbottom KM, Mannering SI, Andrews DM, Voskoboinik I and Trapani JA: Failed CTL/NK cell killing and cytokine hypersecretion are directly linked through prolonged synapse time. J Exp Med. 212:307–317. 2015.PubMed/NCBI View Article : Google Scholar

82 

Voskoboinik I, Thia MC, Fletcher J, Ciccone A, Browne K, Smyth MJ and Trapani JA: Calcium-dependent plasma membrane binding and cell lygis by perforin are mediated through its C2 domain: A critical role for aspartate residues 429, 435, 483, and 485 but not 491. J Biol Chem. 280:8426–8434. 2005.PubMed/NCBI View Article : Google Scholar

83 

Taylor MA, Ward B, Schatzle JD and Bennett M: Perforin- and Fas-dependent mechanisms of natural killer cell-mediated rejection of incompatible bone marrow cell grafts. Eur J Immunol. 32:793–799. 2002.PubMed/NCBI View Article : Google Scholar

84 

Voskoboinik I and Trapani JA: Perforinopathy: A spectrum of human immune disease caused by defective perforin delivery or function. Front Immunol. 4(441)2013.PubMed/NCBI View Article : Google Scholar

85 

Molleran Lee S, Villanueva J, Sumegi J, Zhang K, Kogawa K, Davis J and Filipovich AH: Characterisation of diverse PRF1 mutations leading to decreased natural killer cell activity in North American families with haemophagocytic lymphohistiocytosis. J Med Genet. 41:137–144. 2004.PubMed/NCBI View Article : Google Scholar

86 

Rubin TS, Zhang K, Gifford C, Lane A, Choo S, Bleesing JJ and Marsh RA: Perforin and CD107a testing is superior to NK cell function testing for screening patients for genetic HLH. Blood. 129:2993–2999. 2017.PubMed/NCBI View Article : Google Scholar

87 

Hernandez-Pigeon H, Jean C, Charruyer A, Haure MJ, Titeux M, Tonasso L, Quillet-Mary A, Baudouin C, Charveron M and Laurent G: Human keratinocytes acquire cellular cytotoxicity under UV-B irradiation. Implication of granzyme B and perforin. J Biol Chem. 281:13525–13532. 2006.PubMed/NCBI View Article : Google Scholar

88 

Hernandez-Pigeon H, Jean C, Charruyer A, Haure MJ, Baudouin C, Charveron M, Quillet-Mary A and Laurent G: UVA Induces Granzyme B in Human Keratinocytes through MIF implication in extracellular matrix remodeling. J Biol Chem. 282:8157–8164. 2007.PubMed/NCBI View Article : Google Scholar

89 

Shrestha AC, Wijesundara DK, Masavuli MG, Mekonnen ZA, Gowans EJ and Grubor-Bauk B: Cytolytic perforin as an adjuvant to enhance the immunogenicity of DNA vaccines. Vaccines (Basel). 7(38)2019.PubMed/NCBI View Article : Google Scholar

90 

Gargett T, Grubor-Bauk B, Garrod TJ, Yu W, Miller D, Major L, Wesselingh S, Suhrbier A and Gowans EJ: Induction of antigen-positive cell death by the expression of Perforin, but not DTa, from a DNA vaccine enhances the immune response. Immunol Cell Biol. 92:359–367. 2014.PubMed/NCBI View Article : Google Scholar

91 

Gummow J, Li Y, Yu W, Garrod T, Wijesundara D, Brennan AJ, Mullick R, Voskoboinik I, Grubor-Bauk B and Gowans EJ: A Multiantigenic DNA vaccine that induces broad hepatitis C Virus-specific T-cell responses in mice. J Virol. 89:7991–8002. 2015.PubMed/NCBI View Article : Google Scholar

92 

Wijesundara DK, Yu W, Quah BJC, Eldi P, Hayball JD, Diener KR, Voskoboinik I, Gowans EJ and Grubor-Bauk B: Cytolytic DNA vaccine encoding lytic perforin augments the maturation of- and antigen presentation by-dendritic cells in a time-dependent manner. Sci Rep. 7(8530)2017.PubMed/NCBI View Article : Google Scholar

93 

Escher F, Kühl U, Lassner D, Stroux A, Gross U, Westermann D, Pieske B, Poller W and Schultheiss HP: High perforin-positive cardiac cell infiltration and male sex predict adverse long-term mortality in patients with inflammatory cardiomyopathy. J Am Heart Assoc. 6(e005352)2017.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

August-2022
Volume 24 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pașatu-Cornea A, Ciciu E and Tuță L: Perforin: An intriguing protein in allograft rejection immunology (Review). Exp Ther Med 24: 519, 2022
APA
Pașatu-Cornea, A., Ciciu, E., & Tuță, L. (2022). Perforin: An intriguing protein in allograft rejection immunology (Review). Experimental and Therapeutic Medicine, 24, 519. https://doi.org/10.3892/etm.2022.11446
MLA
Pașatu-Cornea, A., Ciciu, E., Tuță, L."Perforin: An intriguing protein in allograft rejection immunology (Review)". Experimental and Therapeutic Medicine 24.2 (2022): 519.
Chicago
Pașatu-Cornea, A., Ciciu, E., Tuță, L."Perforin: An intriguing protein in allograft rejection immunology (Review)". Experimental and Therapeutic Medicine 24, no. 2 (2022): 519. https://doi.org/10.3892/etm.2022.11446