Apoptosis induced by the lymphocyte effector molecule perforin

https://doi.org/10.1016/j.coi.2007.04.007Get rights and content

Recent progress has been made in discovering structure/function relationships of the cytotoxic lymphocyte pore-forming protein — perforin — and its role in immune regulation. Long recognized as a key component of the granule exocytosis mechanism of lymphocyte-mediated apoptosis and in host defence against a variety of pathogens and tumors, more recent attention has focused upon mutations of the perforin gene in some patients with hemophagocytic lymphohistiocytosis. This clinical picture has prompted further work to uncover the molecular structure of perforin and to define its precise role in immune regulation and immunopathology.

Introduction

Cytotoxic lymphocytes, including cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells, recognize virus-infected or transformed cells and destroy them through perforin-dependent and/or death receptor–death ligand pathways. As such, these pathways represent fundamental mechanisms of both immune surveillance and immunoregulation. Cytotoxic lymphocyte granules contain perforin and many additional pro-apoptotic proteins that are co-secreted to kill target cells. Perforin is encoded by a single copy gene, is highly conserved from fish to primates, and is crucial for the function of the other granule constituents such as granzymes involved in triggering caspase-dependent and caspase-independent target cell death following the formation of an immunological synapse. Mutations and polymorphisms in this effector pathway have been described in mice and humans, and interestingly the phenotypes principally involve defective immune surveillance and/or derangement of lymphoid homeostasis. This review summarizes the most recent and rapid developments in our understanding of the molecular basis of the granule exocytosis effector pathway and its pathophysiological importance.

Section snippets

Developments in the molecular biology of perforin

Granule exocytosis occurs following cytotoxic lymphocyte–target cell synapse formation and involves the polarized trafficking of secretory granules towards the synapse. Granule fusion with the plasma membrane results in the release of perforin and pro-apoptotic serine proteases (granzymes) as well as many other molecules including granulysin and chemokines. Perforin is a pore-forming protein of ∼67 kDa, the expression of which is regulated during lymphocyte differentiation by receptor activation

Developments in the pathophysiology of perforin

Loss of perforin function in mice is associated with immune dysregulation and impaired cytotoxicity, clearly demonstrating the crucial role perforin has as an immune effector molecule [27, 28, 29]. Perforin-deficient mice are viable and healthy under specific pathogen-free conditions, however compromised cellular cytotoxicity is observed upon challenge with certain viruses and they display defects in tumor and transplant rejection. In humans, congenital perforin deficiency accounts for many of

Conclusions

In summary, although there is now no doubt regarding the pivotal importance of lymphocyte perforin in immune response and immune regulation, recent progress in the study of perforin pathophysiology has been relatively slow. In particular, the important discoveries that regulatory lymphocytes express and utilize perforin and that granzyme B might contribute to the death of Th2 cells raise the possibility that interpreting the biology from perforin- or granzyme-deficient hosts might be more

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

PB is supported by a Cancer Research Institute Pre-doctoral Emphasis in Tumor Immunology Scholarship. MJS and JAT are supported by Research Fellowships and a Program Grant from the National Health and Medical Research Council (NHMRC) of Australia. IV receives a career development award from NHMRC. We also thank our colleagues in the Cancer Immunology Program and our collaborators for contributions to many of the findings referred to in this review.

References (73)

  • A.J. Bredemeyer et al.

    Use of protease proteomics to discover granzyme B substrates

    Immunol Res

    (2005)
  • D. Kaiserman et al.

    The major human and mouse granzymes are structurally and functionally divergent

    J Cell Biol

    (2006)
  • M. Muschen et al.

    The origin of CD95-gene mutations in B-cell lymphoma

    Trends Immunol

    (2002)
  • P.A. Mehta et al.

    Perforin polymorphism A91V and susceptibility to B-precursor childhood acute lymphoblastic leukemia: a report from the Children's Oncology Group

    Leukemia

    (2006)
  • J. Loh et al.

    Virgin HWt: natural killer cells utilize both perforin and gamma interferon to regulate murine cytomegalovirus infection in the spleen and liver

    J Virol

    (2005)
  • J.A. Trapani et al.

    Functional significance of the perforin/granzyme cell death pathway

    Nat Rev Immunol

    (2002)
  • U. Zur Stadt et al.

    Mutation spectrum in children with primary hemophagocytic lymphohistiocytosis: molecular and functional analyses of PRF1, UNC13D, STX11, and RAB27A

    Hum Mutat

    (2006)
  • W.J. Grossman et al.

    Human T regulatory cells can use the perforin pathway to cause autologous target cell death

    Immunity

    (2004)
  • M.J. Smyth et al.

    Differential tumor surveillance by natural killer (NK) and NKT cells

    J Exp Med

    (2000)
  • Y. Tang et al.

    Control of immunodeficiency and lymphoproliferation in mouse AIDS: studies of mice deficient in CD8+ T cells or perforin

    J Virol

    (1997)
  • J. Nitcheu et al.

    Perforin-dependent brain-infiltrating cytotoxic CD8+ T lymphocytes mediate experimental cerebral malaria pathogenesis

    J Immunol

    (2003)
  • M.G. Lichtenheld et al.

    Structure and function of human perforin

    Nature

    (1988)
  • E.A. Nalefski et al.

    The C2 domain calcium-binding motif: structural and functional diversity

    Protein Sci

    (1996)
  • R.B. Sutton et al.

    Crystal structure of the cytosolic C2A–C2B domains of synaptotagmin III. Implications for Ca+2-independent SNARE complex interaction

    J Cell Biol

    (1999)
  • R. Uellner et al.

    Perforin is activated by a proteolytic cleavage during biosynthesis which reveals a phospholipid-binding C2 domain

    EMBO J

    (1997)
  • K.N. Balaji et al.

    Surface cathepsin B protects cytotoxic lymphocytes from self-destruction after degranulation

    J Exp Med

    (2002)
  • K. Baran et al.

    Cytotoxic T lymphocytes from cathepsin B-deficient mice survive normally in vitro and in vivo after encountering and killing target cells

    J Biol Chem

    (2006)
  • R. Dressel et al.

    Granzyme-mediated cytotoxicity does not involve the mannose 6-phosphate receptors on target cells

    J Biol Chem

    (2004)
  • C.H. Bird et al.

    Cationic sites on granzyme B contribute to cytotoxicity by promoting its uptake into target cells

    Mol Cell Biol

    (2005)
  • K.A. Browne et al.

    Cytosolic delivery of granzyme B by bacterial toxins: evidence that endosomal disruption, in addition to transmembrane pore formation, is an important function of perforin

    Mol Cell Biol

    (1999)
  • D. Keefe et al.

    Perforin triggers a plasma membrane-repair response that facilitates CTL induction of apoptosis

    Immunity

    (2005)
  • D. Chowdhury et al.

    The exonuclease TREX1 is in the SET complex and acts in concert with NM23-H1 to degrade DNA during granzyme A-mediated cell death

    Mol Cell

    (2006)
  • J.M. Kelly et al.

    Granzyme M mediates a novel form of perforin-dependent cell death

    J Biol Chem

    (2004)
  • M.S. Buzza et al.

    Extracellular matrix remodeling by human granzyme B via cleavage of vitronectin, fibronectin, and laminin

    J Biol Chem

    (2005)
  • F. Andrade et al.

    A novel domain in adenovirus L4-100K is required for stable binding and efficient inhibition of human granzyme B: possible interaction with a species-specific exosite

    Mol Cell Biol

    (2003)
  • D. Kagi et al.

    Cytotoxicity mediated by T cells and natural killer cells is greatly impaired in perforin-deficient mice

    Nature

    (1994)
  • Cited by (114)

    • Structures and functions of the membrane-damaging pore-forming proteins

      2022, Advances in Protein Chemistry and Structural Biology
      Citation Excerpt :

      Perforins on the other hand, are secreted by the cytotoxic T lymphocytes and Natural Killer cells (NK cells) (Podack & Konigsberg, 1984). Perforins generate pores on the virus-infected cells and cancerous host cells (Bolitho, Voskoboinik, Trapani, & Smyth, 2007). Owing to its large pore size, perforins assist in delivering the pro-apoptotic proteases, granzymes, into the target host cells leading to its programmed cell-death (Bird et al., 2005; Metkar et al., 2015; Voskoboinik, Smyth, & Trapani, 2006).

    • Unleashing TNF cytotoxicity to enhance cancer immunotherapy

      2021, Trends in Immunology
      Citation Excerpt :

      In the context of cancer, activation of tumor-specific CTLs involves the binding of their TCR to a tumor peptide antigen presented in the context of MHC-I on the target or antigen-presenting cell, as well as through binding of co-stimulatory receptors to their respective ligands [55]. In the presence of additional cytokine signals, these events lead to the formation of a cytotoxic immunological synapse between the CTL and target cell, resulting in the coordinated release of lytic granules [56], including perforin and granzyme B. Together, these proteins activate apoptotic cascades in target cells and directly cause cell death [5]. In addition, ligation of T cell-associated membrane-bound death ligands, such as Fas ligand (FasL) and TNF-related apoptosis-inducing ligand (TRAIL), to their respective death receptors (DRs) expressed on tumor cells, can also contribute to cytotoxicity [57].

    View all citing articles on Scopus
    View full text