Research report
Murine vesicular monoamine transporter 2: molecular cloning and genomic structure

https://doi.org/10.1016/S0169-328X(97)00116-2Get rights and content

Abstract

The principal brain vesicular monoamine transporter (VMAT2) pumps monoamines including dopamine, norepinephrine, serotonin and histamine from neuronal cytoplasm into synaptic vesicles and is implicated in actions of certain psychostimulants and selective neurotoxins. To improve understanding of this gene and its regulation, and to facilitate study of the roles played by this important molecule in mouse genetic models, we have cloned murine VMAT2 cDNA and genomic sequences. A 4.2-kb mouse VMAT2 cDNA hybridized to a 4.3-kb mRNA expressed chiefly in brainstem. Murine cDNA and genomic DNA analyses reveal an open reading frame of 1551 bp encoding 517 amino acids that display 92, 96 and 60% amino-acid identity with human and rat VMAT2, and rat vesicular acetylcholine transporter sequences, respectively. This open reading frame is distributed over 15 of 16 identified exons, and spans >35 kb of genomic DNA. A major transcriptional initiation site is identified 107 bp 5′ to the translational initiation ATG codon using primer extension/5′ rapid amplification of cDNA ends. Sequences immediately 5′ of this putative transcription start site lack `TATA' or `CATT' boxes, but contain consensus sequences that may bind cAMP response element, Sp1, AP2 and other transcription factors. Identification of these genomic sequences facilitates construction of homologous recombinant mice, provides a template for gene structures in the vesicular transporter family, and identifies sequences elements that could contribute to the specific patterns of regulated VMAT2 expression in monoaminergic neurons.

Introduction

Monoamine neurotransmitters including dopamine, norepinephrine, serotonin and histamine are accumulated into synaptic vesicles by the brain vesicular monoamine transporter (VMAT2) 13, 16, 17, 29, 33. VMAT2 pumps monoamines into vesicles using energy from proton co-transport down the pH gradient generated by the vesicular H+-ATPase 8, 13, 16, 17, 33. VMAT2 mRNA is localized to monoaminergic neurons and VMAT2 protein is largely localized to synaptic vesicular membranes 11, 25, 34, 40, 44. Indeed, VMAT2 is the principal currently identified gene whose expression is restricted to monoaminergic neurons.

VMAT2 function is important for proper packaging of monoamines for quantal, calcium-dependent vesicular release 13, 16, 17, 33. In addition, it may be important for sequestering toxins. The active metabolite, 1-methyl-4-phenyl-phenydium (MPP+), of the selective dopamine neuronal toxin N-methyl-1,2,3,6-tetrahydropyridine (MPTP) kills dopaminergic neurons or other cells that express the plasma membrane dopamine transporter (DAT) and causes a parkinsonian syndrome in vivo 1, 12, 18, 21, 35. Overexpression of VMAT, however, can suppress the toxicity of MPP+ by sequestering the toxin in vesicles away from presumed sites of cytoplasmic mitochondrial damage 24, 25, 26.

Amphetamines cause cytoplasmic release of monoamines from vesicular stores through disruption of the pH gradient across vesicular membranes 37, 39. The balance between this release and VMAT2-mediated vesicular re-uptake may be important for the non-quantal, calcium-independent extracellular dopamine release that modest amphetamine doses can yield, and for the dopaminergic toxicity that high-dose amphetamines can exert, perhaps through cytoplasmic oxidative stresses with dopamine oxidation and free radical production [38].

Recent molecular cloning studies have identified rat and human VMAT2 cDNAs, identifying this transporter as closely related to the VMAT1 cDNA clones expressed largely in adrenal chromaffin cells (VMAT1) and more distantly to the synaptic vesicular acetylcholine transporter 6, 7, 23, 25, 36, 40. However, no study to date has elucidated the structure and function of the VMAT2 gene. To seek insights into regulation of this protein so important for determining vesicular and cytoplasmic monoamine concentrations, to provide candidate sequences for cis acting elements necessary for monoaminergic-specific expression and to allow construction of transgenic mice in which altered expression of VMAT2 could allow better testing of postulated roles for VMAT2 in neurodegenerations and amphetamine actions, we now report elucidation of murine VMAT2 cDNAs and genomic sequences.

Section snippets

Identification of murine VMAT2 hybridization probe

Total RNA was isolated from murine midbrain tissue using RNAsolB™ (TEL-TEST). cDNA was prepared from 1 μg of this RNA using moloney murine leukemia virus (MMLV) reverse transcriptase and conditions suggested by the supplier (Clontech). cDNAs were amplified using the polymerase chain reaction (PCR) and two pairs of oligonucleotide primer sets: F1/R1 (5′-ATGGCCCTGAGCGATCTGGTGCTCTGCG-3′/5′-TGCTGGTAGCCTTGTGTGACTGCCCTCCTGG-3′), and F2/R2

cDNA cloning

Screening of 1×106 recombinant λ cDNA library plaques resulted in the isolation of two hybridizing clones. One of these clones, pCRIImVMAT2-cC contained a 4.2-kb insert. The nucleotide sequence of this clone revealed an open reading frame of 1551 bp, flanked by 35 bp of 5′ and 2.6 kb of 3′ untranslated region that included a polyadenylation signal (Fig. 1). The predicted 517 amino-acid protein showed 96, 92 and 60% amino-acid identify to rat and human VMAT2, and rat acetylcholine vesicular

Discussion

The present report documents sequences of the VMAT2 cDNA and gene of likely importance in identifying structural elements of the gene and its encoded protein, provides an assessment of the likelihood of presence of other closely related genomic sequences, identifies possible sequence elements that might contribute to monoamine-neuron-specific gene expression, and provides genomic elements useful for constructing transgenic mice with VMAT2 gene deletions and/or replacements.

The VMAT2 amino-acid

Acknowledgements

We thank Drs. David J. Vandenbergh and Christopher K. Surratt in our laboratory for helpful advice, Roxann Ingersoll in DNA Analysis Facility, Johns Hopkins University School of Medicine, for sequence analysis, Mary Jane Robinson and Angela Flood for help with the manuscript. We received support from the Intramural Research Program of the National Institute of Drug Abuse.

References (45)

  • A. Merickel et al.

    Transport of histamine by vesicular monoamine t transporter-2

    Neuropharmacology

    (1995)
  • N. Mori et al.

    A cell type-preferred silencer element that controls the neural-specific expression of the SCG10 gene

    Neuron

    (1990)
  • D. Njus et al.

    Biogenetics of secretory vesicles

    Biochem. Biophys. Acta

    (1986)
  • D. Sulzer et al.

    Amphetamine and other psychostimulants reduce pH gradients in midbrain dopaminergic neurons and chromaffin granules: a mechanism of action

    Neuron

    (1990)
  • C.K. Surratt et al.

    A human synaptic vesicle monoamine transporter cDNA predicts posttranslational modifications, reveals chromosome 10 gene localization and identifies TaqI RFLPs

    FEBS Lett.

    (1993)
  • G.R. Uhl

    Neurotransmitter transporters (plus): a promising new gene family

    Trends Neurosci.

    (1992)
  • R.S. Burns et al.

    A primate model of parkinsonism: selective destruction of dopaminergic neurons in the pars compacta of the substantia nigra by N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

    Proc. Natl. Acad. Sci. USA

    (1983)
  • J. Carcamo et al.

    A TATA-like sequence located downstream of the transcription initiation site is required for expression of an RNA polymerase II transcribed gene

    Genes Dev.

    (1990)
  • C. Desnos et al.

    Regulation of the chromaffin granule catecholamine transporter in cultured bovine adrenal medullary cells: stimulus-biosynthesis coupling

    J. Neurochem.

    (1992)
  • J.D. Erickson et al.

    Functional identification and molecular cloning of a human brain vesicle monoamine transporter

    J. Neurochem.

    (1993)
  • J.D. Erickson et al.

    Expression cloning of a reserpine-sensitive vesicular monoamine transporter

    Proc. Natl. Acad. Sci. USA

    (1992)
  • M. Forgac

    Structure and function of vacuolar class of ATP-driven proton pumps

    Physiol. Rev.

    (1989)
  • Cited by (26)

    • N-terminus regulation of VMAT2 mediates methamphetamine-stimulated efflux

      2014, Neuroscience
      Citation Excerpt :

      Unlike the longer PMT N-termini, the hVMAT2 is only 20 AAs in length (Fig. 1). It shares 80% homology with the VMAT1 and similar to the monoamine PMTs the N-terminus is putatively localized to the cytosol (Erickson et al., 1992; Liu et al., 1992a; Erickson and Eiden, 1993; Howell et al., 1994; Takahashi and Uhl, 1997; Duerr et al., 1999). Previous investigations have ascribed regulatory functions to the VMAT2 C-terminus (Krantz et al., 1997; Tan et al., 1998; Waites et al., 2001; Li et al., 2005) and the large luminal-loop domain between TMs 1 and 2 (Ahnert-Hilger et al., 1998, 2003; Holtje et al., 2000; Brunk et al., 2006; Yao and Hersh, 2007).

    • Decreased vesicular monoamine transporter 2 (VMAT2) and dopamine transporter (DAT) function in knockout mice affects aging of dopaminergic systems

      2014, Neuropharmacology
      Citation Excerpt :

      In the extravesicular cytoplasmic compartments of dopaminergic neurons, DA can produce free radicals and/or form adducts with important cellular proteins that can contribute to cellular stress and damage (Uhl, 1998). Extravesicular cytoplasmic DA concentrations are regulated by the serial actions of two transporters: the plasma membrane dopamine transporter (DAT), which mediates neuronal uptake of DA from extracellular spaces into this compartment (Giros et al., 1992; Kilty et al., 1991; Nirenberg et al., 1996b; Shimada et al., 1991; Usdin et al., 1991), and the vesicular monoamine transporter 2 (VMAT2), which translocates DA from this extravesicular cytoplasmic compartment into synaptic vesicles (Erickson et al., 1992; Gonzalez et al., 1994; Liu et al., 1994, 1996, 1992; Merickel et al., 1995; Nirenberg et al., 1996a; Peter et al., 1996, 1995; Roghani et al., 1996; Surratt et al., 1993; Takahashi and Uhl, 1997). Amphetamine-like compounds block VMAT2-mediated transport of DA into synaptic vesicles, elevating extravesicular cytoplasmic DA levels and releasing DA via mechanisms that include DAT-mediated reverse transport.

    • Regulation of Gastric Acid Secretion

      2006, Physiology of the Gastrointestinal Tract
    View all citing articles on Scopus
    View full text