Elsevier

Progress in Neurobiology

Volume 194, November 2020, 101886
Progress in Neurobiology

Original Research Article
A missense point mutation in nerve growth factor (NGFR100W) results in selective peripheral sensory neuropathy

https://doi.org/10.1016/j.pneurobio.2020.101886Get rights and content

Highlights

  • A NGFR100W knockin mouse model of Hereditary Sensory Autonomic Neuropathy V (HSAN V).

  • Homozygous mutant mice showed severe,deficits in peripheral sensory structure and function.

  • Heterozygous mutant mice developed progressive degeneration of small sensory fibers.

  • Heterozygous mutant mice showed no apparent structural/functional changes in the brain.

  • HSAN V is likely resulted from reduced secretion of NGF.

Abstract

The R100W mutation in nerve growth factor is associated with hereditary sensory autonomic neuropathy V in a Swedish family. These patients develop severe loss of perception to deep pain but with apparently normal cognitive functions. To better understand the disease mechanism, we examined a knockin mouse model of HSAN V. The homozygous mice showed significant structural deficits in intra-epidermal nerve fibers (IENFs) at birth. These mice had a total loss of pain perception at ∼2 months of age and often failed to survive to adulthood. Heterozygous mutant mice developed a progressive degeneration of small sensory fibers both behaviorally and functionally: they showed a progressive loss of IENFs starting at the age of 9 months accompanied with progressive loss of perception to painful stimuli such as noxious temperature. Quantitative analysis of lumbar 4/5 dorsal root ganglia revealed a significant reduction in small size neurons, while analysis of sciatic nerve fibers revealed the heterozygous mutant mice had no reduction in myelinated nerve fibers. Significantly, the amount of NGF secreted from mouse embryonic fibroblasts were reduced from both heterozygous and homozygous mice compared to their wild-type littermates. Interestingly, the heterozygous mice showed no apparent structural alteration in the brain: neither the anterior cingulate cortex nor the medial septum including NGF-dependent basal forebrain cholinergic neurons. Accordingly, these animals did not develop appreciable deficits in tests for brain function. Our study has thus demonstrated that the NGFR100W mutation likely affects the structure and function of peripheral sensory neurons.

Introduction

Nerve growth factor (NGF) is a member of the neurotrophic factor family (NTF) that includes brain-derived neurotrophic factor (BDNF), neurotrophin 3 (NT-3) and neurotrophin 4 (NT-4) (Chao, 2003; Chao and Hempstead, 1995; Huang and Reichardt, 2001; Levi-Montalcini, 1987, 2004; Levi-Montalcini et al., 1995). These trophic factors act through two distinct receptors, Trk, the 140 kD tyrosine receptor kinase (TrkA for NGF; TrkB for BDNF, NT-3; TrkC for NT-4) and the 75 kD neurotrophin receptor (p75NTR) to transmit signals in responsive neurons (Bothwell, 1995; Chao and Hempstead, 1995; Kaplan and Miller, 1997). The trophic function of NTFs is largely mediated by Trk, while p75NTR has a more diverse effects on survival, differentiation and death of neurons (Casaccia-Bonnefil et al., 1998; Chao and Hempstead, 1995; Nykjaer et al., 2005).

NGF exerts potent trophic actions on sensory and sympathetic neurons of the peripheral nervous system (PNS)(Hamburger and Levi-Montalcini, 1949) and also regulates the trophic status of striatal and basal forebrain cholinergic neurons (BFCNs) of the central nervous system (CNS)(Conover and Yancopoulos, 1997; Kew et al., 1996; Lehmann et al., 1999; Levi-Montalcini and Hamburger, 1951; Li and Jope, 1995; Svendsen et al., 1994). Given its robust trophic effects, NGF has been investigated for therapeutic properties for treating both PNS and CNS diseases. For example, NGF was explored for treating/preventing degeneration of BFCNs in Alzheimer’s disease (AD) (Blesch and Tuszynski, 1995; Cuello et al., 2010; Eriksdotter Jonhagen et al., 1998; Hefti, 1994; Knusel and Gao, 1996; Koliatsos, 1996; Mufson et al., 2008; Olson, 1993; Rafii et al., 2014; Schindowski et al., 2008; Schulte-Herbruggen et al., 2008; Scott and Crutcher, 1994; Williams et al., 2006). However, some significant issues associated with administration of recombinant NGF such as back pain, injection site hyperalgesia, myalgia, weight loss led to the termination of these trials(Eriksdotter Jonhagen et al., 1998; Hefti, 1994; Knusel and Gao, 1996; Koliatsos, 1996; Olson, 1993; Scott and Crutcher, 1994). Other clinical efforts using NGF for treating diabetic neuropathies, HIV-induced peripheral neuropathies were also terminated due to the extreme side effects of pain(Apfel, 2002; Apfel et al., 1998; Hellweg and Hartung, 1990; Lein, 1995; McArthur et al., 2000; Pradat, 2003; Quasthoff and Hartung, 2001; Rask, 1999; Schifitto et al., 2001; Unger et al., 1998; Walwyn et al., 2006). These adverse effects such as severe pain associated with NGF administration has proven to be a significant roadblock for NGF-based therapies.

Indeed, NGF has also been recognized as a potent mediator of pain (Chuang et al., 2001; Lewin and Mendell, 1993; Lewin et al., 1993; Watanabe et al., 2008). This is further supported by genetic and clinical evidence demonstrating that both TrkA and p75NTR-mediated signaling contributes to NGF-induced hyper-sensitization. For example, hereditary sensory and autonomic neuropathy type IV (HSAN IV) is resulted from recessive mutations in TrkA, these patients display pain insensitivity as well as mental retardation (Indo, 2001, 2002). Furthermore, many TrkA downstream effectors have also been implicated in NGF-mediated nociception: Pharmacological Inhibition of either the extracellular signal-regulated kinases (Erk) or phosphoinositide 3-kinase (PI3K) attenuates NGF-induced hyperalgesia (Zhang and Nicol, 2004). Activation of Phospholipase C (PLCγ) by NGF also potentiates nociceptive ion channels leading to hyperalgesia (Chuang et al., 2001). p75NTR is also involved in NGF-induced hyperalgesia. For example, injecting a p75NTR neutralizing antibody blocked NGF-induced hyperalgesia and NGF-mediated sensitization of action potentials in sensory neurons (Watanabe et al., 2008; Zhang and Nicol, 2004). Ceramide, a p75 downstream effector, is known to increase the number of action potentials in sensory neurons (Zhang et al., 2002, 2006). Nerve injury, axotomy, seizure, or ischemia can all cause an increase in both the expression and axonal transport of p75NTR, thereby contributing to nociception (Zhou et al., 1996) (Roux et al., 1999). p75NTR downstream signaling cascades were responsible for mechanical hyperalgesia following NGF injection (Khodorova et al., 2013). Therefore, both TrkA and p75NTR receptor(s)-mediated signaling pathways play an important role in the pain signaling and function of NGF, although their relative contribution is yet to be defined.

The discovery of NGF mutations in human patients further highlights the importance of NGF in nociception. A homozygous mutation in the NGF gene [680C > A]+[681_682delGG] has been linked to hereditary sensory neuropathy in a consanguineous Arab family(Carvalho et al., 2011). Those affected individuals were completely unable to perceive pain, did not sweat, could not discriminate temperature (Carvalho et al., 2011). In addition, the patients had a chronic immunodeficiency and showed intellectual disabilities (Carvalho et al., 2011).

In a second case, a missense mutation in NGF (661C > T) was discovered in patients in consanguineous Swedish families who suffered from severe loss of deep pain, bone fractures and joint destruction(Einarsdottir et al., 2004). The disorder was classified as HSAN V (Online Mendelian Inheritance in Man (OMIM) # 608654). This particular mutation resulted in a substitution of tryptophan (W) for arginine (R) at Residue 211 in the proNGF polypeptide (pro-NGFR221W that corresponds to Residue 100 in the mature protein: NGFR100W) (Einarsdottir et al., 2004). The mutation is thus referred as NGFR100W herein. Interestingly, unlike HSAN IV patients with recessive TrkA mutation(s) that develop pain insensitivity as well as mental retardation, HSAN V patients suffer from selective loss of pain sensation with normal cognitive function. This suggests that the NGFR100W mutation causes selective loss of pain function, but likely retains intact trophic function (Einarsdottir et al., 2004; Minde et al., 2009, 2004; Minde, 2006; Perini et al., 2016; Sagafos et al., 2016). Therefore, NGFR100W provides an important tool to uncouple the trophic function of NGF from its nociceptive actions (Capsoni, 2014; Capsoni et al., 2011; Cattaneo and Capsoni, 2019; Sung et al., 2018, 2019; Testa et al., 2019a; Yang et al., 2018).

Previous studies have revealed that the R100W mutation might disrupt the processing of proNGF to mature NGF (Larsson et al., 2009). We and others also examined the binding and signaling properties of the mature form of naturally occurring mutant NGF (NGFR100W) and discovered that NGFR100W retained its ability to bind to and signal through TrkA to induce trophic effects, but failed to bind or activate p75NTR (Covaceuszach et al., 2010; Sung et al., 2018). Consistent with these studies, NGFR100E or NGFP61SR100E was also shown to be able to activate TrkA, but not p75NTR signaling (Capsoni et al., 2011; Covaceuszach et al., 2010). Together, these studies have provided strong support for a role of p75NTR in NGF-induced pain function. Failure to activate p75NTR signaling is likely a major reason for loss of pain in HSAN V patients (Capsoni, 2014; Capsoni et al., 2011; Cattaneo and Capsoni, 2019; Sung et al., 2018, 2019; Testa et al., 2019a,b; Yang et al., 2018).

HSAN V is an extremely rare disease that makes it impossible to perform systematic study of signaling mechanism and function of NGFR100W in HSAN V patients. Given that NGF is highly conserved between mice and human, mouse models of HSAN V harboring NGFR100W have provided an important vehicle to carefully examine the effects of NGFR100W on development/ maintenance of both the central nervous system (CNS) and the peripheral nervous system (PNS) in vivo (Testa et al., 2019a; Yang et al., 2018). Using the knock-in mouse model of HSAN V that harboring the mouse NGFR100W allele, we previously reported that mice carrying homozygous NGFR100W alleles (fln/fln) displayed normal embryonic development of major organs (heart, lung, liver, kidney, and spleen) showed normal gene expression of either TrkA or p75NTR receptor (Yang et al., 2018). Interestingly, they exhibited extremely low counts of intra-epidermal nerve fibers (IENFs) at birth (Yang et al., 2018). In a recent study, Testa and colleagues have demonstrated that knocking in a human NGF allele carrying the R100W mutation resulted in a deficit in nociception but apparently with normal learning or memory (Testa et al., 2019b).

In the present study, we further characterized the impact of NGFR100W on the structure and function of the brain and peripheral nervous system in the knock-in mouse model of HSAN V carrying mouse NGFR100W mutation (Yang et al., 2018). We showed that unlike the homozygotes (fln/fln) that often failed to thrive to adulthood, development and survival of the heterozygotes (+/fln) showed no apparent difference from their wt (+/+) littermates. However, the +/fln mice developed a progressive degeneration of small sensory fibers, that were observed both behaviorally and functionally. Interestingly, the +/fln showed no obvious structural alterations in their brain and neuronal populations that included NGF-dependent basal forebrain cholinergic neurons (BFCNs). The +/fln mice did not develop appreciable deficits in learning and memory. Our results are in line with data from limited human studies of NGFR100W carriers (Minde et al., 2009, 2004; Minde, 2006). Mechanistically, we showed the R100W mutation resulted in reduced secretion of mature NGF in primary mouse embryonic fibroblasts (MEFs) cultured from our mutant mice. These results are consistent with previous findings(Larsson et al., 2009; Testa et al., 2019b). We believe these in vivo findings will help to reignite efforts of using NGF as a therapeutic agent, that will lead to the development of ‘painless NGF’ therapies (Capsoni et al., 2011; Cattaneo and Capsoni, 2019; Sung et al., 2019; Testa et al., 2019b).

Section snippets

Ethical statement

All experiments involving the use of laboratory animals have been approved by the Institutional Animal Care and Use Committee of University of California San Diego. Surgical and animal procedures were carried out strictly following the NIH Guide for the Care and Use of Laboratory Animals. In most experiments, both male and female mice were used. When possible, we made every effort in our experiments to measure differences between male and female animals. Mice that were visibly sick, had wounds

The NGFR100W knockin mouse model

To better investigate the impact of NGFR100W mutation, we and other investigators have generated knockin mouse models (Testa et al., 2019a, b; Yang et al., 2018). Testa and colleagues introduced a human NGF allele harboring the R100W mutation to replace mouse endogenous NGF allele (Testa et al., 2019a, b). We simply generated a knockin mouse model of HSAN-V NGFR100W that carried the mouse NGF allele (Yang et al., 2018). Herein, we will refer the wildtype mice as +/+, the heterozygous mutant

Discussion

NGF plays a critical role in supporting the survival and differentiation of specific neuronal populations (Chao, 2003; Huang and Reichardt, 2001; Levi-Montalcini, 1987, 2004; Levi-Montalcini et al., 1995; Levi-Montalcini and Hamburger, 1951; Mobley et al., 1986). Since its discovery, NGF has been extensively explored for its therapeutic potential for treating neurodegenerative diseases (Apfel, 2002; Apfel et al., 1998; Eriksdotter Jonhagen et al., 1998; McArthur et al., 2000; Olson, 1993;

Author contributions

WY, KS, JD and CW designed the study; WY. KS, WX, MJR, ACW, SAS, SF, RKU, SXD, BCG, XO, JR performed the experiments. WY, KS, KJ, NC, RR, JD and CW wrote the paper. All authors discussed the results and commented on the manuscript.

Acknowledgements

We would like to thank Dr William C Mobley for advice and all members of our laboratory for comments and suggestions. We also thank Ms Pauline Hu and Dr. Olga Prikhodko for technical assistance; We would also like Ms. Angela Zeng, Mr. Simon Kim for helping with quantitation of IENFs; This research was supported by grants from: China Postdoctoral Science Foundation (No.2019M662987) to WY; UCSD T32 Neuroplasticity Training Grant to KS; National Natural Science Foundation of China (No.81600920) to

References (95)

  • B. Knusel et al.

    Neurotrophins and Alzheimer’s disease: beyond the cholinergic neurons

    Life Sci.

    (1996)
  • E. Larsson et al.

    Nerve growth factor R221W responsible for insensitivity to pain is defectively processed and accumulates as proNGF

    Neurobiol. Dis.

    (2009)
  • K.F. Lee et al.

    Targeted mutation of the gene encoding the low affinity NGF receptor p75 leads to deficits in the peripheral sensory nervous system

    Cell

    (1992)
  • R. Levi-Montalcini et al.

    Update of the NGF saga

    J. Neurol. Sci.

    (1995)
  • G.R. Lewin et al.

    Nerve growth factor and nociception

    Trends Neurosci.

    (1993)
  • W.C. Mobley et al.

    Nerve growth factor increases choline acetyltransferase activity in developing basal forebrain neurons

    Brain Res.

    (1986)
  • A. Nykjaer et al.

    p75NTR--live or let die

    Curr. Opin. Neurobiol.

    (2005)
  • L. Olson

    NGF and the treatment of Alzheimer’s disease

    Exp. Neurol.

    (1993)
  • F. Paoletti et al.

    A comparative analysis of the structural, functional and biological differences between mouse and human nerve growth factor

    Biochim. Biophys. Acta

    (2015)
  • M.S. Rafii et al.

    A phase1 study of stereotactic gene delivery of AAV2-NGF for Alzheimer’s disease

    Alzheimer’s & dementia: J. Alzheimer’s Assoc.

    (2014)
  • G. Testa et al.

    Cholinergic striatal neurons are increased in HSAN V homozygous mice despite reduced NGF bioavailability

    Biochem. Biophys. Res. Commun.

    (2019)
  • J.W. Unger et al.

    Nerve growth factor (NGF) and diabetic neuropathy in the rat: morphological investigations of the sural nerve, dorsal root ganglion, and spinal cord

    Exp. Neurol.

    (1998)
  • W.M. Walwyn et al.

    HSV-1-mediated NGF delivery delays nociceptive deficits in a genetic model of diabetic neuropathy

    Exp. Neurol.

    (2006)
  • B.J. Williams et al.

    Nerve growth factor in treatment and pathogenesis of Alzheimer’s disease

    Prog. Neurobiol.

    (2006)
  • Y.H. Zhang et al.

    NGF-mediated sensitization of the excitability of rat sensory neurons is prevented by a blocking antibody to the p75 neurotrophin receptor

    Neurosci. Lett.

    (2004)
  • S.C. Apfel et al.

    Recombinant human nerve growth factor in the treatment of diabetic polyneuropathy. NGF Study Group

    Neurology

    (1998)
  • A. Blesch et al.

    Ex vivo gene therapy for Alzheimer’s disease and spinal cord injury

    Clin. Neurosci.

    (1995)
  • M. Bothwell

    Functional interactions of neurotrophins and neurotrophin receptors

    Annu. Rev. Neurosci.

    (1995)
  • S. Capsoni

    From genes to pain: nerve growth factor and hereditary sensory and autonomic neuropathy type V

    Eur. J. Neurosci.

    (2014)
  • S. Capsoni et al.

    Taking pain out of NGF: a "painless" NGF mutant, linked to hereditary sensory autonomic neuropathy type V, with full neurotrophic activity

    PLoS One

    (2011)
  • A.C. Carpenter et al.

    Development of hyperactivity and anxiety responses in dopamine transporter-deficient mice

    Dev. Neurosci.

    (2012)
  • O.P. Carvalho et al.

    A novel NGF mutation clarifies the molecular mechanism and extends the phenotypic spectrum of the HSAN5 neuropathy

    J. Med. Genet.

    (2011)
  • P. Casaccia-Bonnefil et al.

    Neurotrophins: the biological paradox of survival factors eliciting apoptosis

    Cell Death Differ.

    (1998)
  • M.V. Chao

    Neurotrophins and their receptors: a convergence point for many signalling pathways

    Nat. Rev. Neurosci.

    (2003)
  • K.S. Chen et al.

    Disruption of a single allele of the nerve growth factor gene results in atrophy of basal forebrain cholinergic neurons and memory deficits

    J. neurosci. Off. J. Soc. Neurosci.

    (1997)
  • H.H. Chuang et al.

    Bradykinin and nerve growth factor release the capsaicin receptor from PtdIns(4,5)P2-mediated inhibition

    Nature

    (2001)
  • J.C. Conover et al.

    Neurotrophin regulation of the developing nervous system: analyses of knockout mice

    Rev. Neurosci.

    (1997)
  • A.C. Cuello et al.

    Cholinergic involvement in Alzheimer’s disease. A link with NGF maturation and degradation

    J. Mol. Neurosci.

    (2010)
  • D.C. de Andrade et al.

    Beyond neuropathy in hereditary sensory and autonomic neuropathy type V: cognitive evaluation

    Eur. J. Neurol.

    (2008)
  • E. Einarsdottir et al.

    A mutation in the nerve growth factor beta gene (NGFB) causes loss of pain perception

    Hum. Mol. Genet.

    (2004)
  • M. Eriksdotter Jonhagen et al.

    Intracerebroventricular infusion of nerve growth factor in three patients with Alzheimer’s disease

    Dement. Geriatr. Cogn. Disord.

    (1998)
  • Y. Fukui et al.

    Low affinity NGF receptor (p75 neurotrophin receptor) inhibitory antibody reduces pain behavior and CGRP expression in DRG in the mouse sciatic nerve crush model

    J. Orthop. Res.

    (2010)
  • N. Haga et al.

    Hereditary sensory and autonomic neuropathy types IV and V in Japan

    Pediatr. Int.

    (2015)
  • V. Hamburger et al.

    Proliferation, differentiation and degeneration in the spinal ganglia of the chick embryo under normal and experimental conditions

    J. Exp. Zool.

    (1949)
  • F. Hefti

    Development of effective therapy for Alzheimer’s disease based on neurotrophic factors

    Neurobiol. Aging

    (1994)
  • R. Hellweg et al.

    Endogenous levels of nerve growth factor (NGF) are altered in experimental diabetes mellitus: a possible role for NGF in the pathogenesis of diabetic neuropathy

    J. Neurosci. Res.

    (1990)
  • E.J. Huang et al.

    Neurotrophins: roles in neuronal development and function

    Annu. Rev. Neurosci.

    (2001)
  • Cited by (4)

    • Understanding pain perception through genetic painlessness diseases: The role of NGF and proNGF

      2021, Pharmacological Research
      Citation Excerpt :

      Their early postnatal lethality could be rescued by NGFWT administration [69], reminiscent of the phenotype caused by the complete deletion of the NGF gene [11]. A reduced hNGFR221W bioavailability, due to defects in its secretion [26,41,70], represents the main explanation for the lethality of hNGFR221W/R221W mice. Early lethality is also observed in another mouse model for HSAN V, harbouring the R221W mutation in the context of the mouse Ngf gene [71].

    1

    These authors contributed equally

    View full text