Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Bruce L. Patton is active.

Publication


Featured researches published by Bruce L. Patton.


Current Pharmaceutical Design | 2009

Developmental and Pathogenic Mechanisms of Basement Membrane Assembly

Bruce L. Patton

Basement membranes are sheet-like cell-adherent extracellular matrices that serve as cell substrata and solid-phase agonists, contributing to tissue organization, stability and differentiation. These matrices are assembled as polymers of laminins and type IV collagens that are tethered to nidogens and proteoglycans. They bind to cell surface molecules that include signal-transducing receptors such as the integrins and dystroglycan and form attachments to adjacent connective tissues. The cell receptors, in turn, provide links between the matrix and underlying cytoskeleton. Genetic diseases of basement membrane and associated components, collectively the basement membrane zone, disrupt the extracellular matrix and/or its linkages to affect nerve, muscle, skin, kidney and other tissues. These diseases can arise due to a loss of matrix integrity, adhesion strength and/or receptor-mediated signaling. An understanding of the mechanisms of basement membrane zone assembly and resulting structure can provide insights into the development of normal tissues and the pathogenic mechanisms that underlie diverse disorders.


Nature Neuroscience | 2001

Properly formed but improperly localized synaptic specializations in the absence of laminin alpha4.

Bruce L. Patton; Jeanette M. Cunningham; Jill Thyboll; Jarkko Kortesmaa; Håkan Westerblad; Lars Edström; Karl Tryggvason; Joshua R. Sanes

Precise apposition of pre- to postsynaptic specializations is required for optimal function of chemical synapses, but little is known about how it is achieved. At the skeletal neuromuscular junction, active zones (transmitter release sites) in the nerve terminal lie directly opposite junctional folds in the postsynaptic membrane. Few active zones or junctional folds form in mice lacking the laminin β2 chain, which is normally concentrated in the synaptic cleft. β2 and the broadly expressed γ1 chain form heterotrimers with α chains, three of which, α2, α4 and α5, are present in the synaptic cleft. Thus, α2β2γ1, α4β2γ1 and α5β2γ1 heterotrimers are all lost in β2 mutants. In mice lacking laminin α4, active zones and junctional folds form in normal numbers, but are not precisely apposed to each other. Thus, formation and localization of synaptic specializations are regulated separately, and α4β2γ1 (called laminin-9) is critical in the latter process.


The Journal of Neuroscience | 2007

β1 Integrins Control the Formation of Cell Chains in the Adult Rostral Migratory Stream

Richard Belvindrah; Sabine Hankel; John R. Walker; Bruce L. Patton; Ulrich Müller

The subventricular zone (SVZ) of the lateral ventricle is the major site of neurogenesis in the adult brain. Neuroblasts that are born in the SVZ migrate as chains along the rostral migratory stream (RMS) to the olfactory bulb. Little is known about the mechanisms that control interactions between neuroblasts during their migration. Here we show that migrating neuroblasts express β1integrins and that the integrin ligand laminin is localized to cell chains. Using genetically modified mice and time-lapse video recordings of SVZ explants, we demonstrate that β1 integrins and laminin promote the formation of cell chains. Laminin also induces the aggregation of purified neuroblasts. We conclude that the formation of cell chains in the RMS is controlled in part by β1 integrins via binding to laminin. In addition, we provide evidence that β1 class integrins are required for the maintenance of the glial tubes and that defects in the glial tubes lead to the ectopic migration of neuroblasts into the surrounding tissue.


Nature | 1998

Synaptic laminin prevents glial entry into the synaptic cleft.

Bruce L. Patton; Arlene Y. Chiu; Joshua R. Sanes

Presynaptic and postsynaptic membranes directly oppose each other at chemical synapses, minimizing the delay in transmitting information across the synaptic cleft. Extrasynaptic neuronal surfaces, in contrast, are almost entirely covered by processes from glial cells. The exclusion of glial cells from the synaptic cleft, and the long-term stability of synapses, presumably result in large part from the tight adhesion between presynaptic and postsynaptic elements,. Here we show that there is another requirement for synaptic maintenance: glial cells of the skeletal neuromuscular synapse, Schwann cells, are actively inhibited from entering the synaptic cleft between the motor nerve terminal and the muscle fibre. One inhibitory component is laminin 11, a heterotrimeric glycoprotein that is concentrated in the synaptic cleft. Regulation of an inhibitory interaction between glial cells and synaptic cleft components may contribute to synaptic rearrangements, and loss of this inhibition may underlie the loss of synapses that results from injury to the postsynaptic cell.


PLOS Biology | 2009

β1 Integrin Maintains Integrity of the Embryonic Neocortical Stem Cell Niche

Karine Loulier; Justin D. Lathia; Véronique Marthiens; Jenne Relucio; Mohamed R. Mughal; Sung-Chun Tang; Turhan Coksaygan; Peter E. Hall; Srinivasulu Chigurupati; Bruce L. Patton; Holly Colognato; Mahendra S. Rao; Mark P. Mattson; Tarik F. Haydar; Charles ffrench-Constant

IInteractions between laminins and integrin receptors hold neural stem cells in place at the ventricular surface of embryonic brain. Transient disruption leads to abnormal stem cell divisions and permanent cortical malformation.


Journal of Cell Biology | 2005

Coordinate control of axon defasciculation and myelination by laminin-2 and -8

Dongren Yang; Jesse Bierman; Yukie S. Tarumi; Yong Ping Zhong; Reshma Rangwala; Thomas M. Proctor; Yuko Miyagoe-Suzuki; Shin'ichi Takeda; Jeffrey H. Miner; Larry S. Sherman; Bruce G. Gold; Bruce L. Patton

Schwann cells form basal laminae (BLs) containing laminin-2 (Ln-2; heterotrimer α2β1γ1) and Ln-8 (α4β1γ1). Loss of Ln-2 in humans and mice carrying α2-chain mutations prevents developing Schwann cells from fully defasciculating axons, resulting in partial amyelination. The principal pathogenic mechanism is thought to derive from structural defects in Schwann cell BLs, which Ln-2 scaffolds. However, we found loss of Ln-8 caused partial amyelination in mice without affecting BL structure or Ln-2 levels. Combined Ln-2/Ln-8 deficiency caused nearly complete amyelination, revealing Ln-2 and -8 together have a dominant role in defasciculation, and that Ln-8 promotes myelination without BLs. Transgenic Ln-10 (α5β1γ1) expression also promoted myelination without BL formation. Rather than BL structure, we found Ln-2 and -8 were specifically required for the increased perinatal Schwann cell proliferation that attends myelination. Purified Ln-2 and -8 directly enhanced in vitro Schwann cell proliferation in collaboration with autocrine factors, suggesting Lns control the onset of myelination by modulating responses to mitogens in vivo.


The Journal of Neuroscience | 2010

Quiescence and activation of stem and precursor cell populations in the subependymal zone of the mammalian brain are associated with distinct cellular and extracellular matrix signals.

Ilias Kazanis; Justin D. Lathia; Eric Raborn; Ruiqian Wan; Mohamed R. Mughal; D. Mark Eckley; Takako Sasaki; Bruce L. Patton; Mark P. Mattson; Karen K. Hirschi; Mary E. Dickinson; Charles ffrench-Constant

The subependymal zone (SEZ) of the lateral ventricles is one of the areas of the adult brain where new neurons are continuously generated from neural stem cells (NSCs), via rapidly dividing precursors. This neurogenic niche is a complex cellular and extracellular microenvironment, highly vascularized compared to non-neurogenic periventricular areas, within which NSCs and precursors exhibit distinct behavior. Here, we investigate the possible mechanisms by which extracellular matrix molecules and their receptors might regulate this differential behavior. We show that NSCs and precursors proceed through mitosis in the same domains within the SEZ of adult male mice—albeit with NSCs nearer ependymal cells—and that distance from the ventricle is a stronger limiting factor for neurogenic activity than distance from blood vessels. Furthermore, we show that NSCs and precursors are embedded in a laminin-rich extracellular matrix, to which they can both contribute. Importantly, they express differential levels of extracellular matrix receptors, with NSCs expressing low levels of α6β1 integrin, syndecan-1, and lutheran, and in vivo blocking of β1 integrin selectively induced the proliferation and ectopic migration of precursors. Finally, when NSCs are activated to reconstitute the niche after depletion of precursors, expression of laminin receptors is upregulated. These results indicate that the distinct behavior of adult NSCs and precursors is not necessarily regulated via exposure to differential extracellular signals, but rather via intrinsic regulation of their interaction with their microenvironment.


The Journal of Comparative Neurology | 2007

Patterns of laminins and integrins in the embryonic ventricular zone of the CNS

Justin D. Lathia; Bruce L. Patton; D. Mark Eckley; Tim Magnus; Mohamed R. Mughal; Takako Sasaki; Maeve A. Caldwell; Mahendra S. Rao; Mark P. Mattson; Charles ffrench-Constant

The extracellular matrix (ECM) provides both a physical framework and a microenvironment that supplies instructive signals from the earliest stages of multicellular development. As a first step toward understanding the role of the ECM in regulating the behavior of neural stem cells (NSCs), here we show the localization of laminins, a heterotrimeric family of ECM molecules expressed in many different stem cell microenvironments, and their corresponding receptors in the embryonic murine ventricular zone (VZ) within which the NSCs undergo symmetrical and asymmetrical divisions required for cortical development. In addition to the presence of laminins containing both the α2 and α4 chains, we find distinct patterns of ECM receptor expression in the VZ and in the overlying cortex. Neural stem cells derived from the VZ express high levels of the integrin laminin receptor α6β1. At developmental stages at which NSCs undergo asymmetrical divisions, integrin β1 was unevenly distributed in some mitotic pairs at the ventricular wall. These results suggest a significant role in the regulation of NSC fate for laminin/integrin signaling within the microenvironment of the VZ and provide a framework for future molecular and cellular analyses of the role of the ECM in neural development. J. Comp. Neurol. 505:630–643, 2007.


Journal of The American Society of Nephrology | 2006

A Hypomorphic Mutation in the Mouse Laminin α5 Gene Causes Polycystic Kidney Disease

M. Brendan Shannon; Bruce L. Patton; Scott J. Harvey; Jeffrey H. Miner

Extracellular matrix abnormalities have been found in both human and animal models of polycystic kidney disease (PKD). A new mouse PKD model has been produced through insertion of a PGKneo cassette in an intron of the gene that encodes laminin alpha5 (Lama5), a major tubular and glomerular basement membrane component that is important for glomerulogenesis and ureteric bud branching. Lama5neo represents a hypomorphic allele as a result of aberrant splicing. Lama5neo/neo mice exhibit PKD, proteinuria, and death from renal failure by 4 wk of age. This contrasts with mice that totally lack Lama5, which die in utero with multiple developmental defects. At 2 d of age, Lama5neo/neo mice exhibited mild proteinuria and microscopic cystic transformation. By 2 wk, cysts were grossly apparent in cortex and medulla, involving both nephron and collecting duct segments. Tubular basement membranes seemed to form normally, and early cyst basement membranes showed normal ultrastructure but developed marked thickening as cysts enlarged. Overall, Lama5 protein levels were severely reduced as a result of mRNA frameshift caused by exon skipping. This was accompanied by aberrant accumulation of laminin-332 (alpha3beta3gamma2; formerly called laminin-5) in some cysts, as also observed in human PKD. This constitutes the first evidence that a primary defect in an extracellular matrix component can cause PKD.


The Journal of Neuroscience | 2009

Regulation of radial glial survival by signals from the meninges

Randor Radakovits; Claudia S. Barros; Richard Belvindrah; Bruce L. Patton; Ulrich Müller

Radial glial cells (RGCs) in the developing cerebral cortex are progenitors for neurons and glia, and their processes serve as guideposts for migrating neurons. So far, it has remained unclear whether RGC processes also control the function of RGCs more directly. Here, we show that RGC numbers and cortical size are reduced in mice lacking β1 integrins in RGCs. TUNEL stainings and time-lapse video recordings demonstrate that β1-deficient RGCs processes detach from the meningeal basement membrane (BM) followed by apoptotic death of RGCs. Apoptosis is also induced by surgical removal of the meninges. Finally, mice lacking the BM components laminin α2 and α4 show defects in the attachment of RGC processes at the meninges, a reduction in cortical size, and enhanced apoptosis of RGC cells. Our findings demonstrate that attachment of RGC processes at the meninges is important for RGC survival and the control of cortical size.

Collaboration


Dive into the Bruce L. Patton's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffrey H. Miner

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Arlene Y. Chiu

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jeanette M. Cunningham

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark P. Mattson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mohamed R. Mughal

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

D. Mark Eckley

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mahendra S. Rao

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge