Trends in Cell Biology
Volume 18, Issue 11, November 2008, Pages 560-574
Journal home page for Trends in Cell Biology

Review
Feature Review
Breaching the basement membrane: who, when and how?

https://doi.org/10.1016/j.tcb.2008.08.007Get rights and content

The basement membrane (BM), a specialized network of extracellular matrix macromolecules, surrounds epithelial, endothelial, muscle, fat and nerve cells. During development, immune surveillance and disease states ranging from cancer to fibrosis, host cells penetrate the BM by engaging tissue-invasive programs, the identity of which remain largely undefined. Although it is commonly assumed that all cells employ similar mechanisms to cross BM barriers, accumulating evidence indicates that cells might selectively mobilize protease-dependent or -independent invasion programs. New data indicate that protease-dependent transmigration is largely reliant on a group of membrane-anchored metalloenzymes, termed the membrane-type matrix metalloproteinases, which irreversibly remodel BM structure. By contrast, mechanisms that enable protease-independent transmigration remain undefined and potentially involve the reversible disassembly of the BM network. Further characterization of the molecular mechanisms underlying BM transmigration should provide important insights into pathophysiologic tissue remodeling events and also enable the development of novel therapeutics.

Introduction

Early in development, animals ranging from flies to humans direct the embryonic epithelium to orchestrate the organization of an extracellular, supramolecular network of proteins, glycoproteins and proteoglycans, termed the basement membrane (BM) 1, 2, 3. This conglomerate of structural macromolecules coalesces to form a dense, 100–300 nm-thick lamina that underlies all epithelia and, in higher organisms, ensheaths endothelial cells, nerves, smooth muscle cells and adipocytes 1, 4, 5. The assembled BM provides adherent cells with structural support and functional cues by virtue of its biomechanical properties, display of adhesion receptor ligands and repertoire of matrix-bound growth factors 4, 6. With a pore size in the order of 50 nm, only small molecules are able to passively diffuse across this thin, but structurally rugged, barrier 6, 7, 8. Nonetheless, normal cells are able to traffic freely and rapidly across BMs by activating tissue-invasive programs during morphogenesis and immune surveillance 9, 10, 11, 12, 13. Furthermore, in a repetitive theme familiar to biologists, cell populations participating in pathologic events such as cancer can inappropriately co-opt ‘normal’ BM-transmigration programs to dire and, most often, lethal consequences by driving the metastatic process [14].

The transmigration of cells across the BM has an unquestionably important role in normal and neoplastic events and has been the subject of thousands of reports – not to mention innumerable reviews – in the literature 1, 2, 3, 6, 14. Nevertheless, it probably comes as a surprise that the mechanisms that enable cells to cross this structural barrier remain largely unknown and the subject of considerable debate. Whereas the ability of a migrating cell to perforate the BM has been almost uniformly ascribed to proteolytic events, >500 proteinases are encoded within the mammalian genome, thus complicating efforts to identify a subset of crucial, matrix-degrading enzymes [15]. As such, using various model constructs designed to recapitulate BM structure in vitro, most investigators have, over the past 20 years, accepted a largely circumstantial premise that secreted proteinases directly regulate transmigration in both physiologic and pathologic states 16, 17, 18. Very recently, however, evidence has begun to accumulate indicating that accepted dogma might now be ripe for revisiting 1, 19, 20, 21. New insights into BM assembly and structure have raised serious concerns regarding the use of most of the in vitro models used for analyzing invasion 22, 23, 24. Furthermore, in vivo studies of mice harboring inactivating mutations of proteinases commonly implicated as the ‘usual suspects’ in BM transmigration seldom seem to exhibit frank defects in BM invasion-associated events 25, 26, 27, 28. Instead, recent studies support the contention that a small subset of membrane-anchored metalloproteases assumes a previously unrecognized role in this process [21]. Although we would be pleased to inform readers that solutions to all queries regarding BM invasion programs lie within this perspective, this is far from the case and continued efforts are required to build a definitive model of BM transmigration. Rather, it is our intent to highlight existing conundrums and caveats in the field, to pose possible solutions as to the means by which normal and neoplastic cells traverse BM barriers and to outline experimental systems in which these hypotheses might be evaluated and tested in rigorous fashion.

Section snippets

BM structure

Comprising >50 distinct macromolecules, the predominant components of BMs are intertwined meshworks of polymeric laminin and type IV collagen (see Glossary) 2, 3, 4, 6. Distinct from all other BM constituents, only laminin and type IV collagen are able to self-assemble into polymers [4]. Until recently, most models of BM organization assumed that type IV collagen serves as the major scaffolding upon which the laminin network is deposited. Newer studies indicate, however, that laminin polymers

When do cells cross BMs?

Multiple cell types traverse BM barriers in the course of developmental, inflammatory, fibrotic and neoplastic processes (Figure 2). During mammalian development, the early embryo consists of two apposed layers of epithelial cells. With the inception of gastrulation, genesis of the third germ layer (the mesoderm) occurs when epithelial cells at the primitive streak adopt mesenchymal (i.e. fibroblast-like) characteristics and penetrate the underlying BM to populate the intervening space 11, 12,

Mechanisms of BM transmigration: an introduction

To probe the mechanisms underlying BM transmigration, it must first be asked: what are the properties of BMs and the associated invasive cell populations that influence the transmigration process? From a structural perspective, the migrating cell will be confronted with a semi-permeable, type-IV-collagen-rich barrier, the pore size of which is dictated by both extracellular matrix (ECM) density and crosslinking 5, 21, 43. In turn, the degree to which the cell can deform its cytoplasm and

Breaking down the door: protease-dependent BM transmigration

Evidence for proteolytic BM destruction during transmigration is supported by the observation that tissue-invasive events associated with developmental and disease states are characterized frequently by irreversible changes in BM structure 1, 12, 13, 21, 38, 39. During development in worms, flies and mammals, BM effacement is commonly observed at sites of invasion (e.g. within the vulva in C. elegans and the primitive streak in mammals) 1, 19, 20. Similarly, BM discontinuities have been

Opening the door: non-proteolytic BM transmigration

Clearly, proteinases, especially MT-MMPs, can function as crucial regulators of transmigration by degrading BM-associated structural barriers and generating a conduit through which normal and malignant epithelial cells can pass. By contrast, are there any BM-transmigration events wherein proteolysis is dispensable?

Recently, non-proteolytic means of cell invasion have been reported using Matrigel as a BM surrogate 103, 130, 131. Carcinoma cells are capable of invading Matrigel matrices using a

Concluding remarks and future perspectives

Despite the obvious strengths of in vitro models, the full repertoire of proteolytic and non-proteolytic systems available to normal or neoplastic cells might only be accessible in vivo. As such, additional insights into the role of MT-MMPs in BM transmigration will be gleaned from further analyses of gene-targeted mice. Whereas many developmental BM-invasive events are intact in both Mmp14 and Mmp16 (MT3-MMP) mutant mice 165, 166, current studies indicate that the expression of either MT1-MMP,

Glossary

Anchor cell
a specialized cell in C. elegans that invades the vulval epithelium and crosses the underlying BM during normal worm development.
Basement membrane
a specialized form of extracellular matrix comprised of an interwoven mixture of type IV collagen, laminins, nidogen and sulfated proteoglycans that lies beneath epithelial and endothelial cells and surrounds muscles, nerves, adipocytes and smooth muscle cells.
Branching morphogenesis
during development and angiogenesis, the formation of a

References (173)

  • B. Wu

    Wnt signaling induces matrix metalloproteinase expression and regulates T cell transmigration

    Immunity

    (2007)
  • R.M. Kuntz et al.

    Neutrophil motility in extracellular matrix gels: mesh size and adhesion affect speed of migration

    Biophys. J.

    (1997)
  • V. Pillay

    The urokinase plasminogen activator receptor as a gene therapy target for cancer

    Trends Biotechnol.

    (2007)
  • S. Schenk et al.

    Tales from the crypt[ic] sites of the extracellular matrix

    Trends Cell Biol.

    (2003)
  • X. Fu

    Activation and silencing of matrix metalloproteinases

    Semin. Cell Dev. Biol.

    (2008)
  • J.A. Eble

    The recognition sites of the integrins α1β1 and α2β1 within collagen IV are protected against gelatinase A attack in the native protein

    J. Biol. Chem.

    (1996)
  • A.C. Wheatcroft

    Evidence of in situ stability of the type IV collagen triple helix in human inflammatory bowel disease using a denaturation specific epitope antibody

    Matrix Biol.

    (1999)
  • A.R. Mackay

    Studies on the ability of 65-kDa and 92-kDa tumor cell gelatinases to degrade type IV collagen

    J. Biol. Chem.

    (1990)
  • J.D. Mott

    Nonenzymatic glycation of type IV collagen and matrix metalloproteinase susceptibility

    Kidney Int.

    (1997)
  • D. Kajimura

    Non-helical type IV collagen polypeptides in human placenta

    Biochem. Biophys. Res. Commun.

    (2004)
  • T.H. Vu

    MMP-9/gelatinase B is a key regulator of growth plate angiogenesis and apoptosis of hypertrophic chondrocytes

    Cell

    (1998)
  • K. Inoue

    A crucial role for matrix metalloproteinase 2 in osteocytic canalicular formation and bone metabolism

    J. Biol. Chem.

    (2006)
  • R.A. Dean et al.

    Proteomics discovery of metalloproteinase substrates in the cellular context by iTRAQ labeling reveals a diverse MMP-2 substrate degradome

    Mol. Cell. Proteomics

    (2007)
  • B. Azzarelli et al.

    Amniotic basement membrane: a barrier to neutrophil invasion

    Am. J. Obstet. Gynecol.

    (1987)
  • H.K. Kleinman et al.

    Matrigel: basement membrane matrix with biological activity

    Semin. Cancer Biol.

    (2005)
  • I. Virtanen

    Laminin α1-chain shows a restricted distribution in epithelial basement membranes of fetal and adult human tissues

    Exp. Cell Res.

    (2000)
  • S. Even-Ram et al.

    Cell migration in 3D matrix

    Curr. Opin. Cell Biol.

    (2005)
  • N. Koshikawa

    Membrane-type matrix metalloproteinase-1 (MT1-MMP) is a processing enzyme for human laminin γ 2 chain

    J. Biol. Chem.

    (2005)
  • J. Khoshnoodi

    Mammalian collagen IV

    Microsc. Res. Tech.

    (2008)
  • B.G. Hudson

    Alport’s syndrome, Goodpasture’s syndrome, and type IV collagen

    N. Engl. J. Med.

    (2003)
  • J. Candiello

    Biomechanical properties of native basement membranes

    FEBS J.

    (2007)
  • R. Kalluri

    Basement membranes: structure, assembly and role in tumour angiogenesis

    Nat. Rev. Cancer

    (2003)
  • G.A. Abrams

    Nanoscale topography of the corneal epithelial basement membrane and Descemet’s membrane of the human

    Cornea

    (2000)
  • G.A. Abrams

    Nanoscale topography of the basement membrane underlying the corneal epithelium of the rhesus macaque

    Cell Tissue Res.

    (2000)
  • A.R. Huber et al.

    Disruption of the subendothelial basement membrane during neutrophil diapedesis in an in vitro construct of a blood vessel wall

    J. Clin. Invest.

    (1989)
  • K. Ley

    Getting to the site of inflammation: the leukocyte adhesion cascade updated

    Nat. Rev. Immunol.

    (2007)
  • C. Viebahn

    Morphology of incipient mesoderm formation in the rabbit embryo: a light- and retrospective electron-microscopic study

    Acta Anat. (Basel)

    (1995)
  • C. Viebahn

    Epithelio-mesenchymal transformation during formation of the mesoderm in the mammalian embryo

    Acta Anat. (Basel)

    (1995)
  • G. Christofori

    New signals from the invasive front

    Nature

    (2006)
  • X.S. Puente

    Human and mouse proteases: a comparative genomic approach

    Nat. Rev. Genet.

    (2003)
  • L.A. Liotta

    Metastatic potential correlates with enzymatic degradation of basement membrane collagen

    Nature

    (1980)
  • J. Hu

    Matrix metalloproteinase inhibitors as therapy for inflammatory and vascular diseases

    Nat. Rev. Drug Discov.

    (2007)
  • A. Page-McCaw

    Matrix metalloproteinases and the regulation of tissue remodelling

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • A. Srivastava

    Basement membrane remodeling is essential for Drosophila disc eversion and tumor invasion

    Proc. Natl. Acad. Sci. U. S. A.

    (2007)
  • K. Hotary

    A cancer cell metalloprotease triad regulates the basement membrane transmigration program

    Genes Dev.

    (2006)
  • M.E. Than

    The 1.9-A crystal structure of the noncollagenous (NC1) domain of human placenta collagen IV shows stabilization via a novel type of covalent Met-Lys cross-link

    Proc. Natl. Acad. Sci. U. S. A.

    (2002)
  • S.E. Perez

    Matrix metalloproteinases 2 and 9 are dispensable for pancreatic islet formation and function in vivo

    Diabetes

    (2005)
  • P. Baluk

    Matrix metalloproteinase-2 and -9 expression increases in Mycoplasma-infected airways but is not required for microvascular remodeling

    Am. J. Physiol. Lung Cell. Mol. Physiol.

    (2004)
  • T. Itoh

    The role of matrix metalloproteinase-2 and matrix metalloproteinase-9 in antibody-induced arthritis

    J. Immunol.

    (2002)
  • S. Li

    Laminin-sulfatide binding initiates basement membrane assembly and enables receptor signaling in Schwann cells and fibroblasts

    J. Cell Biol.

    (2005)
  • Cited by (366)

    View all citing articles on Scopus
    View full text