Survey
Hemophagocytic lymphohistiocytosis (HLH): A heterogeneous spectrum of cytokine-driven immune disorders

https://doi.org/10.1016/j.cytogfr.2014.10.001Get rights and content

Highlights

  • HLH comprises a broad spectrum of disorders that all present with a cytokine storm.

  • Diverse animal models of HLH have enhanced our understanding of this disease.

  • Models of primary HLH appoint a major role for IFN-γ in disease pathogenesis.

  • TNF-α, IL-6, IL-10 and IL-18 are also cytokines of interest in design of treatments.

  • Targeting hyperactive T cells or DCs may represent an alternative therapeutic option.

Abstract

Hemophagocytic lymphohistiocytosis (HLH) comprises a group of life-threatening immune disorders classified into primary or secondary HLH. The former is caused by mutations in genes involved in granule-mediated cytotoxicity, the latter occurs in a context of infections, malignancies or autoimmune/autoinflammatory disorders. Both are characterized by systemic inflammation, severe cytokine storms and immune-mediated organ damage. Despite recent advances, the pathogenesis of HLH remains incompletely understood. Animal models resembling different subtypes of HLH are therefore of great value to study this disease and to uncover novel treatment strategies. In this review, all known animal models of HLH will be discussed, highlighting findings on cell types, cytokines and signaling pathways involved in disease pathogenesis and extrapolating therapeutic implications for the human situation.

Introduction

Hemophagocytic lymphohistiocytosis (HLH) is a rare, life-threatening disorder that was first described by Scott and Robb-Smith in 1939 [1]. It is characterized by severe systemic inflammation caused by uncontrolled proliferation and activation of lymphocytes and macrophages, secreting large amounts of cytokines, creating a so-called cytokine storm. The hallmark of HLH is the emergence of hemophagocytosis, a process in which histiocytes actively engulf blood cells and their precursors. Table 1 provides a comprehensive overview of symptoms that are most commonly observed in HLH [2], [3], [4], together with their application in the current diagnostic criteria [5].

Several subtypes of HLH are distinguished on the basis of current knowledge concerning their etiology and pathogenesis, and are classified as either primary or secondary HLH (Table 2). Primary HLH is an autosomal recessive, monogenic disorder, caused by loss-of-function mutations in genes involved in the cytotoxic function of natural killer (NK) cells and CD8+ T cells. The affected proteins and their role in granule exocytosis are depicted in Fig. 1. Secondary HLH is defined by a similar clinical syndrome but is assumed to lack a known genetic basis. This form of HLH is thought to occur in the context of an underlying immunological condition, such as malignancy or an autoimmune or autoinflammatory disorder. Secondary HLH associated with the latter two is also referred to as ‘macrophage activation syndrome’ (MAS). Clinical manifestations in both primary and secondary HLH are often precipitated by an infectious trigger, including viral, bacterial, fungal and protozoan infections, and can possibly be facilitated by immune-modulating therapies [2], [3], [6], [7], [8]. Taken together, HLH comprises a heterogeneous spectrum of etiologically different disorders (Fig. 2), which impedes early recognition of the syndrome, proper diagnosis and timely treatment of the diverse patients [3], [8], [9], [10].

Despite recent advances, the pathogenic mechanisms behind the different subtypes of HLH remain incompletely understood, which hampers the development of effective therapies for all patients. Therapeutic regimens are often empirically based, and patient responses are highly variable. Mortality rates can raise up to 70% in certain subtypes [11]. This accentuates the need for ongoing research. Specific animal models resembling different subtypes of HLH are of great value to study the pathogenesis of this perplexing disease and to uncover novel treatment options. In this paper all known animal models, covering the full spectrum of primary as well as secondary HLH, will be comprehensively reviewed. A summary of all models can be found in Table 3. Findings on cell types, cytokines and signaling pathways playing a role in pathogenesis will be discussed, together with their potential relevance and therapeutic implications for the human situation.

Section snippets

Animal models for primary HLH

The ongoing identification of mutations in different genes playing a role in granule-mediated cytotoxicity in primary HLH patients has driven the development of animal models specific for this subtype of HLH. Mice have been genetically engineered so as to mirror the mutations found in patients, in order to decipher the importance of these genes in disease pathogenesis. Most of these mutant mice do not spontaneously develop HLH but are predisposed to progress to an HLH-like syndrome after an

Spectrum of primary HLH phenotypes: comparative analysis of animal models

A comparative analysis of the different animal models for primary HLH reveals a wide spectrum of disease severity. Not all models reflect the full-blown disease seen in patients. Models of XLP, in particular, fail to clarify how HLH can emerge in XLP1 or XLP2. One hypothesis relates the variable predisposition to HLH in different patients, and in different animal models, to differences in the cytotoxic function. The residual cytotoxic function of a patient with primary HLH is determined by the

Animal models for secondary HLH

The clinical and laboratory manifestations of secondary HLH are identical to that of primary HLH, but no homozygous defects in the cytotoxic function are detectable. Nevertheless, decreased numbers of NK cells, low NK cell activity or reduced perforin expression in cytotoxic cells can be found in patients with secondary HLH [74], [75], [76]. Patients with MAS in particular may even possess heterozygous mutations in genes involved in the cytolytic pathway, blurring the thin line between primary

Therapies in animal models of HLH: implications for the treatment of patients?

Currently, treatment of HLH is predominantly based on the therapeutic guidelines of the HLH-2004 protocol, which prescribes chemo-immunotherapy such as etoposide, dexamethasone, cyclosporine A, intrathecal methotrexate and corticosteroids together with appropriate supportive care. Hematopoietic stem cell transplantation is advised for patients with primary HLH or recurrent disease [5]. However, these approaches are not equally effective in all patients, often resulting in empirical therapy for

Final considerations

The development of very diverse animal models for primary and secondary HLH have greatly enhanced our understanding of these complex syndromes. As discussed in this review, inflammatory cytokines are significantly increased in the large majority of animal models that investigated them, endorsing the importance of a cytokine storm in disease pathogenesis. IFN-γ has been studied in most models and was reported to be consistently increased in all but one model. This is indicative of a major role

Acknowledgements

The authors thank Prof. Alfons Billiau and Dr. Anneleen Avau for critical revision of the manuscript. This work was supported by grants from the Agency for Innovation by Science and Technology (IWT), the Regional Government of Flanders (GOA program), the Fund for Scientific Research Flanders (FWO-Vlaanderen) and the Interuniversity Attraction Poles (IAP). E.B. received a fellowship from the IWT. The authors declare no competing financial interests.

Ellen Brisse obtained her master's degree in Bioscience Engineering and is currently working as a PhD student at the Laboratory of Immunobiology of the Rega Institute (University of Leuven, Belgium). As a fellow of the Agency for Innovation by Science and Technology (IWT), she is investigating the role of pro-inflammatory cytokines, specific cell types and viral infection in mouse models of herpesvirus-associated hemophagocytic lymphohistiocytosis and macrophage activation syndrome complicating

References (150)

  • S.L.H. Van Dommelen et al.

    Perforin and granzymes have distinct roles in defensive immunity and immunopathology

    Immunity

    (2006)
  • M. Chen et al.

    Critical role for perforin and Fas-dependent killing of dendritic cells in the control of inflammation

    Blood

    (2012)
  • J. Feldmann et al.

    Munc13-4 is essential for cytolytic granules fusion and is mutated in a form of familial hemophagocytic lymphohistiocytosis (FHL3)

    Cell

    (2003)
  • P. Krebs et al.

    Disruption of MyD88 signaling suppresses hemophagocytic lymphohistiocytosis in mice

    Blood

    (2011)
  • T. Kögl et al.

    Hemophagocytic lymphohistiocytosis in syntaxin-11-deficient mice: T-cell exhaustion limits fatal disease

    Blood

    (2013)
  • E.K. Novak et al.

    The regulation of platelet-dense granules by Rab27a in the ashen mouse, a model of Hermansky–Pudlak and Griscelli syndromes, is granule-specific and dependent on genetic background

    Blood

    (2002)
  • B. Jessen et al.

    Subtle differences in CTL cytotoxicity determine susceptibility to hemophagocytic lymphohistiocytosis in mice and humans with Chediak–Higashi syndrome

    Blood

    (2011)
  • H.-C. Chuang et al.

    Epstein-Barr virus LMP1 inhibits the expression of SAP gene and upregulates Th1 cytokines in the pathogenesis of hemophagocytic syndrome

    Blood

    (2005)
  • J.M. Rumble et al.

    Phenotypic differences between mice deficient in XIAP and SAP, two factors targeted in X-linked lymphoproliferative syndrome (XLP)

    Cell Immunol

    (2009)
  • C. Rivat et al.

    SAP gene transfer restores cellular and humoral immune function in a murine model of X-linked lymphoproliferative disease

    Blood

    (2013)
  • S. Crotty et al.

    Hypogammaglobulinemia and exacerbated CD8 T-cell-mediated immunopathology in SAP-deficient mice with chronic LCMV infection mimics human XLP disease

    Blood

    (2006)
  • R.A. Marsh et al.

    XIAP deficiency: a unique primary immunodeficiency best classified as X-linked familial hemophagocytic lymphohistiocytosis and not as X-linked lymphoproliferative disease

    Blood

    (2010)
  • C.E. Terrell et al.

    Mixed hematopoietic or T cell chimerism above a minimal threshold restores perforin-dependent immune regulation in perforin-deficient mice

    Blood

    (2013)
  • A. Grom et al.

    Natural killer cell dysfunction in patients with systemic-onset rheumatoid arthritis and macrophage activation syndrome

    J Pediatr

    (2003)
  • K. Kogawa et al.

    Perforin expression in cytotoxic lymphocytes from patients with hemophagocytic lymphohistiocytosis and their family members

    Blood

    (2002)
  • M. Zhang et al.

    Genetic defects in cytolysis in macrophage activation syndrome

    Curr Rheumatol Rep

    (2014)
  • M. Tiab et al.

    Haemophagocytic syndrome associated with infections

    Baillieres Best Pract Res Clin Haematol

    (2000)
  • K. Sato et al.

    A novel animal model of Epstein–Barr virus-associated hemophagocytic lymphohistiocytosis in humanized mice

    Blood

    (2011)
  • J.-I. Henter et al.

    Cytotoxic therapy for severe avian influenza A (H5N1) infection

    Lancet

    (2006)
  • Y. Kasahara et al.

    Cell type specific infection of Epstein–Barr virus (EBV) in EBV-associated hemophagocytic lymphohistiocytosis and chronic active EBV infection

    Crit Rev Oncol Hematol

    (2002)
  • K. Hayashi et al.

    Rabbit model for human EBV-associated hemophagocytic syndrome (HPS), Sequential autopsy analysis and characterization of IL-2 dependent cell lines established from Herpesvirus papio-induced fatal rabbit lymphoproliferative disease with HPS

    Am J Pathol

    (2003)
  • W.-C. Hsieh et al.

    Emergence of anti-red blood cell antibodies triggers red cell phagocytosis by activated macrophages in a rabbit model of Epstein–Barr virus-associated hemophagocytic syndrome

    Am J Pathol

    (2007)
  • J. Naessens

    Bovine trypanotolerance: a natural ability to prevent severe anaemia and haemophagocytic syndrome?

    Int J Parasitol

    (2006)
  • R. Scott et al.

    Histiocytic medullary reticulocytosis

    Lancet

    (1939)
  • G.E. Janka

    Familial and acquired hemophagocytic lymphohistiocytosis

    Eur J Pediatr

    (2007)
  • J.I. Henter et al.

    HLH-2004: diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis

    Pedriatr Blood Cancer

    (2007)
  • J.W. Verbsky et al.

    Hemophagocytic lymphohistiocytosis: diagnosis, pathophysiology, treatment, and future perspectives

    Ann Med

    (2006)
  • E. Sieni et al.

    Familial hemophagocytic lymphohistiocytosis: a model for understanding the human machinery of cellular cytotoxicity

    Cell Mol Life Sci

    (2012)
  • C. Gholam et al.

    Familial haemophagocytic lymphohistiocytosis: advances in the genetic basis, diagnosis and management

    Clin Exp Immunol

    (2011)
  • S. Deane et al.

    Macrophage activation syndrome in autoimmune disease

    Int Arch Allergy Immunol

    (2010)
  • M. Ramos-Casals et al.

    Adult haemophagocytic syndrome

    Lancet

    (2013)
  • C.M. Walsh et al.

    Immune function in mice lacking the perforin gene

    Proc Natl Acad Sci USA

    (1994)
  • D. Kägi et al.

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

    Nature

    (1994)
  • A. Müllbacher et al.

    Perforin is essential for control of ectromelia virus but not related poxviruses in mice

    J Virol

    (1999)
  • B. Jessen et al.

    Graded defects in cytotoxicity determine severity of hemophagocytic lymphohistiocytosis in humans and mice

    Front Immunol

    (2013)
  • D. Binder et al.

    Aplastic anemia rescued by exhaustion of cytokine-secreting CD8+ T cells in persistent infection with lymphocytic choriomeningitis virus

    J Exp Med

    (1998)
  • M. Matloubian et al.

    A role for perforin in downregulating T-cell responses during chronic viral infection

    J Virol

    (1999)
  • J. Pachlopnik Schmid et al.

    Neutralization of IFNgamma defeats haemophagocytosis in LCMV-infected perforin- and Rab27a-deficient mice

    EMBO Mol Med

    (2009)
  • T.S. Johnson et al.

    Etoposide selectively ablates activated T cells to control the immunoregulatory disorder hemophagocytic lymphohistiocytosis

    J Immunol

    (2014)
  • E.E. Zoller et al.

    Hemophagocytosis causes a consumptive anemia of inflammation

    J Exp Med

    (2011)
  • Cited by (145)

    View all citing articles on Scopus

    Ellen Brisse obtained her master's degree in Bioscience Engineering and is currently working as a PhD student at the Laboratory of Immunobiology of the Rega Institute (University of Leuven, Belgium). As a fellow of the Agency for Innovation by Science and Technology (IWT), she is investigating the role of pro-inflammatory cytokines, specific cell types and viral infection in mouse models of herpesvirus-associated hemophagocytic lymphohistiocytosis and macrophage activation syndrome complicating rheumatic diseases.

    Carine H. Wouters is a professor at the Faculty of Medicine at the University of Leuven and a pediatric rheumatologist, affiliated to the University Hospital of Leuven and Hôpital Necker-Enfants Malades in Paris. She participates in multiple international projects to develop guidelines for diagnoses and treatments of juvenile arthritis and macrophage activation syndrome, including clinical trials with anti-cytokine therapies.

    Patrick Matthys is professor in Immunology at the University of Leuven and a research director at the Rega Institute. He earned his PhD in 1993 with experimental work demonstrating the key role of IFN-γ in cytokine release syndromes and in eliciting tumor-associated cachexia. Matthys now continues conducting research at the Rega Institute, focusing on the pathogenesis of autoimmune diseases, in particular autoimmune arthritis, and autoinflammatory syndromes, i.e. systemic juvenile idiopathic arthritis and macrophage activation syndrome. The complex role of IFN-γ in the pathogenesis of these diseases remains a preferred subject of his investigations, together with in-depth analysis of interactions with other cytokines, chemokines and inflammatory mediators.

    1

    Tel.: +32 16 33 73 86; fax: +32 16 33 73 40.

    2

    Tel.: +32 16 34 38 43; fax: +32 16 34 38 42.

    3

    These authors contributed equally to this work.

    View full text