Network


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

Hotspot


Dive into the research topics where Maria Z. Kounnas is active.

Publication


Featured researches published by Maria Z. Kounnas.


Cell | 1995

LDL receptor-related protein, a multifunctional ApoE receptor, binds secreted β-amyloid precursor protein and mediates its degradation

Maria Z. Kounnas; Robert D. Moir; G. William Rebeck; Ashley I. Bush; W. Scott Argraves; Rudolph E. Tanzi; Bradley T. Hyman; Dudley K. Strickland

The secreted form of beta-amyloid precursor protein (APP) containing the Kunitz proteinase inhibitor (KPI) domain, also called protease nexin II, is internalized and degraded by cells. We show that the low density lipoprotein (LDL) receptor-related protein (LRP) is responsible for the endocytosis of secreted APP. APPs770 degradation is inhibited by an LRP antagonist called the receptor-associated protein (RAP) and by LRP antibodies and is greatly diminished in fibroblasts genetically deficient in LRP. APPs695, which lacks the KPI domain, is a poor LRP ligand. Since LRP also binds apolipoprotein E (apoE)-enriched lipoproteins and inheritance of the epsilon 4 allele of the apoE gene is a risk factor for Alzheimers disease (AD), these data link in a single metabolic pathway two molecules strongly implicated in the pathophysiology of AD.


Journal of Clinical Investigation | 2000

Modulation of amyloid β-protein clearance and Alzheimer’s disease susceptibility by the LDL receptor–related protein pathway

David E. Kang; Claus U. Pietrzik; Larry Baum; Nathalie Chevallier; David E. Merriam; Maria Z. Kounnas; Steven L. Wagner; Juan C. Troncoso; Claudia H. Kawas; Robert Katzman; Edward H. Koo

Susceptibility to Alzheimers disease (AD) is governed by multiple genetic factors. Remarkably, the LDL receptor-related protein (LRP) and its ligands, apoE and alpha2M, are all genetically associated with AD. In this study, we provide evidence for the involvement of the LRP pathway in amyloid deposition through sequestration and removal of soluble amyloid beta-protein (Abeta). We demonstrate in vitro that LRP mediates the clearance of both Abeta40 and Abeta42 through a bona fide receptor-mediated uptake mechanism. In vivo, reduced LRP expression is associated with LRP genotypes and is correlated with enhanced soluble Abeta levels and amyloid deposition. Although LRP has been proposed to be a clearance pathway for Abeta, this work provides the first in vivo evidence that the LRP pathway may modulate Abeta deposition and AD susceptibility by regulating the removal of soluble Abeta.


The FASEB Journal | 1995

LDL receptor-related protein: a multiligand receptor for lipoprotein and proteinase catabolism.

Dudley K. Strickland; Maria Z. Kounnas; W S Argraves

The accumulation of excessive choles‐terol‐rich lipoproteins within vascular cells, the pro‐liferation of vascular cells, and fibrin deposition are hallmark features of atherosclerosis. Evidence accu‐mulated over the past few years supports the hypothesis that one member of the LDL receptor family, the low density lipoprotein receptor‐related protein (LRP), affects the dynamics of each of these processes. LRP is expressed in several vascular cell types, including smooth muscle cells, and in macrophages, and is also expressed in these cells in atherosclerotic lesions. This receptor is a large endocytotic receptor that mediates the catabolism of a number of molecules known to be important in vascular biology, including apolipoprotein E‐ and lipoprotein lipase‐enriched lipoproteins, thrombospondin, and plasminogen activators. The capacity of LRP to mediate lipoprotein catabolism may be a factor in the development of the lesion by contributing to the formation of foam cells. LRP has recently been shown to mediate the catabolism of thrombospondin, a molecule that has potent biological effects on cells of the vasculature. The regulation of its extracellular accumulation by LRP might modulate the dynamic processes of tissue remodeling associated with the response to vascular injury. In addition, LRP regulates the expression of plasmin activity by directly binding and mediating the cellular internalization of urokinase‐ and tissue‐type plasminogen activators. The cellular removal of these two enzymes decreases the local profibrinolytic potential, possibly leading to a thrombotic state at lesion rites.—Strickland, D. K., Kounnas, M. Z., Argraves, W. S. LDL receptor‐related protein: a multiligand receptor for lipoprotein and proteinase catabolism. FASEBJ. 9, 890‐898 (1995)


Neuron | 2010

Modulation of γ-Secretase Reduces β-Amyloid Deposition in a Transgenic Mouse Model of Alzheimer's Disease

Maria Z. Kounnas; Anne M. Danks; Soan Cheng; Curtis Tyree; Elizabeth J. Ackerman; Xulun Zhang; Kwangwook Ahn; Phuong D. Nguyen; Dan Comer; Long Mao; Chengzhi Yu; David Pleynet; Paul J. Digregorio; Gonul Velicelebi; Kenneth A. Stauderman; William T. Comer; William C. Mobley; Yueming Li; Sangram S. Sisodia; Rudolph E. Tanzi; Steven L. Wagner

Alzheimers disease (AD) is characterized pathologically by the abundance of senile plaques and neurofibrillary tangles in the brain. We synthesized over 1200 novel gamma-secretase modulator (GSM) compounds that reduced Abeta(42) levels without inhibiting epsilon-site cleavage of APP and Notch, the generation of the APP and Notch intracellular domains, respectively. These compounds also reduced Abeta(40) levels while concomitantly elevating levels of Abeta(38) and Abeta(37). Immobilization of a potent GSM onto an agarose matrix quantitatively recovered Pen-2 and to a lesser degree PS-1 NTFs from cellular extracts. Moreover, oral administration (once daily) of another potent GSM to Tg 2576 transgenic AD mice displayed dose-responsive lowering of plasma and brain Abeta(42); chronic daily administration led to significant reductions in both diffuse and neuritic plaques. These effects were observed in the absence of Notch-related changes (e.g., intestinal proliferation of goblet cells), which are commonly associated with repeated exposure to functional gamma-secretase inhibitors (GSIs).


Journal of Biological Chemistry | 2009

Alzheimer Disease Aβ Production in the Absence of S-Palmitoylation-dependent Targeting of BACE1 to Lipid Rafts

Kulandaivelu S. Vetrivel; Xavier Meckler; Ying Chen; Phuong D. Nguyen; Nabil G. Seidah; Robert Vassar; Philip C. Wong; Masaki Fukata; Maria Z. Kounnas; Gopal Thinakaran

Alzheimer disease β-amyloid (Aβ) peptides are generated via sequential proteolysis of amyloid precursor protein (APP) by BACE1 and γ-secretase. A subset of BACE1 localizes to cholesterol-rich membrane microdomains, termed lipid rafts. BACE1 processing in raft microdomains of cultured cells and neurons was characterized in previous studies by disrupting the integrity of lipid rafts by cholesterol depletion. These studies found either inhibition or elevation of Aβ production depending on the extent of cholesterol depletion, generating controversy. The intricate interplay between cholesterol levels, APP trafficking, and BACE1 processing is not clearly understood because cholesterol depletion has pleiotropic effects on Golgi morphology, vesicular trafficking, and membrane bulk fluidity. In this study, we used an alternate strategy to explore the function of BACE1 in membrane microdomains without altering the cellular cholesterol level. We demonstrate that BACE1 undergoes S-palmitoylation at four Cys residues at the junction of transmembrane and cytosolic domains, and Ala substitution at these four residues is sufficient to displace BACE1 from lipid rafts. Analysis of wild type and mutant BACE1 expressed in BACE1 null fibroblasts and neuroblastoma cells revealed that S-palmitoylation neither contributes to protein stability nor subcellular localization of BACE1. Surprisingly, non-raft localization of palmitoylation-deficient BACE1 did not have discernible influence on BACE1 processing of APP or secretion of Aβ. These results indicate that post-translational S-palmitoylation of BACE1 is not required for APP processing, and that BACE1 can efficiently cleave APP in both raft and non-raft microdomains.


Journal of Clinical Investigation | 2009

NSAIDs prevent, but do not reverse, neuronal cell cycle reentry in a mouse model of Alzheimer disease

Kiran Bhaskar; Maria Z. Kounnas; Steven L. Wagner; Yan Yang; Bruce T. Lamb; Karl Herrup

Ectopic cell cycle events (CCEs) mark vulnerable neuronal populations in human Alzheimer disease (AD) and are observed early in disease progression. In transgenic mouse models of AD, CCEs are found before the onset of beta-amyloid peptide (Abeta) deposition to form senile plaques, a hallmark of AD. Here, we have demonstrated that alterations in brain microglia occur coincidently with the appearance of CCEs in the R1.40 transgenic mouse model of AD. Furthermore, promotion of inflammation with LPS at young ages in R1.40 mice induced the early appearance of neuronal CCEs, whereas treatment with 2 different nonsteroidal antiinflammatory drugs (NSAIDs) blocked neuronal CCEs and alterations in brain microglia without altering amyloid precursor protein (APP) processing and steady-state Abeta levels. In addition, NSAID treatment of older R1.40 animals prevented new neuronal CCEs, although it failed to reverse existing ones. Retrospective human epidemiological studies have identified long-term use of NSAIDs as protective against AD. Prospective clinical trials, however, have failed to demonstrate a similar benefit. Our use of CCEs as an outcome measure offers fresh insight into this discrepancy and provides important information for future clinical trials, as it suggests that NSAID use in human AD may need to be initiated as early as possible to prevent disease progression.


Journal of Biological Chemistry | 2006

Ubiquilin 1 Modulates Amyloid Precursor Protein Trafficking and Aβ Secretion

Mikko Hiltunen; Alice Lu; Anne V. Thomas; Donna M. Romano; Minji Kim; Phill B. Jones; Zhongcong Xie; Maria Z. Kounnas; Steven L. Wagner; Oksana Berezovska; Bradley T. Hyman; Giuseppina Tesco; Lars Bertram; Rudolph E. Tanzi

Ubiquilin 1 (UBQLN1) is a ubiquitin-like protein, which has been shown to play a central role in regulating the proteasomal degradation of various proteins, including the presenilins. We recently reported that DNA variants in UBQLN1 increase the risk for Alzheimer disease, by influencing expression of this gene in brain. Here we present the first assessment of the effects of UBQLN1 on the metabolism of the amyloid precursor protein (APP). For this purpose, we employed RNA interference to down-regulate UBQLN1 in a variety of neuronal and non-neuronal cell lines. We demonstrate that down-regulation of UBQLN1 accelerates the maturation and intracellular trafficking of APP, while not interfering with α-, β-, or γ-secretase levels or activity. UBQLN1 knockdown increased the ratio of APP mature/immature, increased levels of full-length APP on the cell surface, and enhanced the secretion of sAPP (α- and β-forms). Moreover, UBQLN1 knockdown increased levels of secreted Aβ40 and Aβ42. Finally, employing a fluorescence resonance energy transfer-based assay, we show that UBQLN1 and APP come into close proximity in intact cells, independently of the presence of the presenilins. Collectively, our findings suggest that UBQLN1 may normally serve as a cytoplasmic “gatekeeper” that may control APP trafficking from intracellular compartments to the cell surface. These findings suggest that changes in UBQLN1 steady-state levels affect APP trafficking and processing, thereby influencing the generation of Aβ.


Annals of the New York Academy of Sciences | 1994

The α2-Macroglobulin Receptor/Low Density Lipoprotein Receptor-Related Protein and the Receptor-Associated Protein

Suzanne E. Williams; Maria Z. Kounnas; Kelley M. Argraves; W. Scott Argraves; Dudley K. Strickland

a*-Macroglobulin (azM) is a proteinase inhibitor that inhibits all four classes of proteinase. The mechanism of inhibition is unique; aZM is cleaved by the proteinase in a region termed the “bait region” and then undergoes conformational changes that activate internal thiol ester bonds, providing sites for covalent attachment of proteinases and other molecules. These conformational changes also expose receptorbinding domains on the a*M-proteinase complex that mediate its interaction with a specific cell surface receptor that is responsible for removing these complexes from the circulation. The receptor responsible for a*M-proteinase clearance was isolated by ligand affinity chromatography from human placenta tissue’,* and from liver.’ Sequencing studies revealed that the a2M receptor is identical to low density lipoprotein (LDL) receptor-related protein (LRP)!.’ The purified receptor contains a 5 15kDa heavy chain and an 85-kDa light chain that are noncovalently associated. A 39-kDa polypeptide copurifies with the receptor’,* and has been termed the receptorassociated protein (RAP).6 In this report, we will review the structure of LRP, the numerous ligands that are now known to bind to LRP, and the potential function of LRP in their metabolism, as well as review the structure of the receptor-associated protein and its potential function in vivo.


Biofutur | 1997

Modulators of expression and function of LRP in Alzheimer's disease

Dudley K. Strickland; Bradley T. Hyman; Maria Z. Kounnas; Robert D. Moir; Rudolph E. Tanzi; G. William Rebeck

The present invention relates to methods and compositions for preventing the endocytosis and cellular internalization of integral membrane amyloid β-precursor protein (APP) and its subsequent catabolism by blocking or interfering with the association or binding of APP with members of the low density lipoprotein receptor family.


Journal of Biological Chemistry | 2010

Mutation Analysis of the Presenilin 1 N-terminal Domain Reveals a Broad Spectrum of γ-Secretase Activity toward Amyloid Precursor Protein and Other Substrates

Ping Gong; Kulandaivelu S. Vetrivel; Phuong D. Nguyen; Xavier Meckler; Haipeng Cheng; Maria Z. Kounnas; Steven L. Wagner; Angèle Parent; Gopal Thinakaran

The γ-secretase protein complex executes the intramembrane proteolysis of amyloid precursor protein (APP), which releases Alzheimer disease β-amyloid peptide. In addition to APP, γ-secretase also cleaves several other type I membrane protein substrates including Notch1 and N-cadherin. γ-Secretase is made of four integral transmembrane protein subunits: presenilin (PS), nicastrin, APH1, and PEN2. Multiple lines of evidence indicate that a heteromer of PS-derived N- and C-terminal fragments functions as the catalytic subunit of γ-secretase. Only limited information is available on the domains within each subunit involved in the recognition and recruitment of diverse substrates and the transfer of substrates to the catalytic site. Here, we performed mutagenesis of two domains of PS1, namely the first luminal loop domain (LL1) and the second transmembrane domain (TM2), and analyzed PS1 endoproteolysis as well as the catalytic activities of PS1 toward APP, Notch, and N-cadherin. Our results show that distinct residues within LL1 and TM2 domains as well as the length of the LL1 domain are critical for PS1 endoproteolysis, but not for PS1 complex formation with nicastrin, APH1, and PEN2. Furthermore, our experimental PS1 mutants formed γ-secretase complexes with distinct catalytic properties toward the three substrates examined in this study; however, the mutations did not affect PS1 interaction with the substrates. We conclude that the N-terminal LL1 and TM2 domains are critical for PS1 endoproteolysis and the coordination between the putative substrate-docking site and the catalytic core of the γ-secretase.

Collaboration


Dive into the Maria Z. Kounnas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gonul Velicelebi

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

W. Scott Argraves

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kenneth A. Stauderman

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge