Network


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

Hotspot


Dive into the research topics where Maximillian A. Rogers is active.

Publication


Featured researches published by Maximillian A. Rogers.


Proceedings of the National Academy of Sciences of the United States of America | 2010

ACAT1 gene ablation increases 24(S)-hydroxycholesterol content in the brain and ameliorates amyloid pathology in mice with AD

Elena Y. Bryleva; Maximillian A. Rogers; Catherine C. Y. Chang; Floyd Buen; Brent T. Harris; Estelle Rousselet; Nabil G. Seidah; Salvatore Oddo; Frank M. LaFerla; Thomas A. Spencer; William F. Hickey; Ta-Yuan Chang

Cholesterol metabolism has been implicated in the pathogenesis of several neurodegenerative diseases, including the abnormal accumulation of amyloid-β, one of the pathological hallmarks of Alzheimer disease (AD). Acyl-CoA:cholesterol acyltransferases (ACAT1 and ACAT2) are two enzymes that convert free cholesterol to cholesteryl esters. ACAT inhibitors have recently emerged as promising drug candidates for AD therapy. However, how ACAT inhibitors act in the brain has so far remained unclear. Here we show that ACAT1 is the major functional isoenzyme in the mouse brain. ACAT1 gene ablation (A1−) in triple transgenic (i.e., 3XTg-AD) mice leads to more than 60% reduction in full-length human APPswe as well as its proteolytic fragments, and ameliorates cognitive deficits. At 4 months of age, A1− causes a 32% content increase in 24-hydroxycholesterol (24SOH), the major oxysterol in the brain. It also causes a 65% protein content decrease in HMG-CoA reductase (HMGR) and a 28% decrease in sterol synthesis rate in AD mouse brains. In hippocampal neurons, A1− causes an increase in the 24SOH synthesis rate; treating hippocampal neuronal cells with 24SOH causes rapid declines in hAPP and in HMGR protein levels. A model is provided to explain our findings: in neurons, A1− causes increases in cholesterol and 24SOH contents in the endoplasmic reticulum, which cause reductions in hAPP and HMGR protein contents and lead to amelioration of amyloid pathology. Our study supports the potential of ACAT1 as a therapeutic target for treating certain forms of AD.


Human Molecular Genetics | 2012

A novel mouse model of Niemann–Pick type C disease carrying a D1005G-Npc1 mutation comparable to commonly observed human mutations

Robert A. Maue; Robert W. Burgess; Bing Wang; Christine M. Wooley; Kevin L. Seburn; Marie T. Vanier; Maximillian A. Rogers; Catherine C. Y. Chang; Ta-Yuan Chang; Brent T. Harris; David J. Graber; Carlos A. A. Penatti; Donna M. Porter; Benjamin S. Szwergold; Leslie P. Henderson; John W. Totenhagen; Theodore P. Trouard; Ivan A. Borbon; Robert P. Erickson

We have identified a point mutation in Npc1 that creates a novel mouse model (Npc1(nmf164)) of Niemann-Pick type C1 (NPC) disease: a single nucleotide change (A to G at cDNA bp 3163) that results in an aspartate to glycine change at position 1005 (D1005G). This change is in the cysteine-rich luminal loop of the NPC1 protein and is highly similar to commonly occurring human mutations. Genetic and molecular biological analyses, including sequencing the Npc1(spm) allele and identifying a truncating mutation, confirm that the mutation in Npc1(nmf164) mice is distinct from those in other existing mouse models of NPC disease (Npc1(nih), Npc1(spm)). Analyses of lifespan, body and spleen weight, gait and other motor activities, as well as acoustic startle responses all reveal a more slowly developing phenotype in Npc1(nmf164) mutant mice than in mice with the null mutations (Npc1(nih), Npc1(spm)). Although Npc1 mRNA levels appear relatively normal, Npc1(nmf164) brain and liver display dramatic reductions in Npc1 protein, as well as abnormal cholesterol metabolism and altered glycolipid expression. Furthermore, histological analyses of liver, spleen, hippocampus, cortex and cerebellum reveal abnormal cholesterol accumulation, glial activation and Purkinje cell loss at a slower rate than in the Npc1(nih) mouse model. Magnetic resonance imaging studies also reveal significantly less demyelination/dysmyelination than in the null alleles. Thus, although prior mouse models may correspond to the severe infantile onset forms of NPC disease, Npc1(nmf164) mice offer many advantages as a model for the late-onset, more slowly progressing forms of NPC disease that comprise the large majority of human cases.


Journal of Clinical Investigation | 2016

Sortilin mediates vascular calcification via its recruitment into extracellular vesicles

Claudia Goettsch; Joshua D. Hutcheson; Masanori Aikawa; Hiroshi Iwata; Tan Pham; Anders Nykjaer; Mads Kjolby; Maximillian A. Rogers; Thomas Michel; Manabu Shibasaki; Sumihiko Hagita; Rafael Kramann; Daniel J. Rader; Peter Libby; Sasha Singh; Elena Aikawa

Vascular calcification is a common feature of major cardiovascular diseases. Extracellular vesicles participate in the formation of microcalcifications that are implicated in atherosclerotic plaque rupture; however, the mechanisms that regulate formation of calcifying extracellular vesicles remain obscure. Here, we have demonstrated that sortilin is a key regulator of smooth muscle cell (SMC) calcification via its recruitment to extracellular vesicles. Sortilin localized to calcifying vessels in human and mouse atheromata and participated in formation of microcalcifications in SMC culture. Sortilin regulated the loading of the calcification protein tissue nonspecific alkaline phosphatase (TNAP) into extracellular vesicles, thereby conferring its calcification potential. Furthermore, SMC calcification required Rab11-dependent trafficking and FAM20C/casein kinase 2-dependent C-terminal phosphorylation of sortilin. In a murine model, Sort1-deficiency reduced arterial calcification but did not affect bone mineralization. Additionally, transfer of sortilin-deficient BM cells to irradiated atherosclerotic mice did not affect vascular calcification, indicating a primary role of SMC-derived sortilin. Together, the results of this study identify sortilin phosphorylation as a potential therapeutic target for ectopic calcification/microcalcification and may clarify the mechanism that underlies the genetic association between the SORT1 gene locus and coronary artery calcification.


Journal of Biological Chemistry | 2015

Deficiency in the Lipid Exporter ABCA1 Impairs Retrograde Sterol Movement and Disrupts Sterol Sensing at the Endoplasmic Reticulum

Yoshio Yamauchi; Noriyuki Iwamoto; Maximillian A. Rogers; Sumiko Abe-Dohmae; Toyoshi Fujimoto; Catherine C. Y. Chang; Masato Ishigami; Takuma Kishimoto; Toshihide Kobayashi; Kazumitsu Ueda; Koichi Furukawa; Ta-Yuan Chang; Shinji Yokoyama

Background: Little is known about how mammalian cells modulate retrograde sterol transport for cellular cholesterol homeostasis. Results: ABCA1 deficiency impairs endocytic retrograde sterol transport to the endoplasmic reticulum and activates the SREBP-2 pathway. Conclusion: ABCA1 participates in bidirectional sterol movement at the plasma membrane to regulate cellular cholesterol homeostasis. Significance: A novel function of ABCA1 is identified. Cellular cholesterol homeostasis involves sterol sensing at the endoplasmic reticulum (ER) and sterol export from the plasma membrane (PM). Sterol sensing at the ER requires efficient sterol delivery from the PM; however, the macromolecules that facilitate retrograde sterol transport at the PM have not been identified. ATP-binding cassette transporter A1 (ABCA1) mediates cholesterol and phospholipid export to apolipoprotein A-I for the assembly of high density lipoprotein (HDL). Mutations in ABCA1 cause Tangier disease, a familial HDL deficiency. Several lines of clinical and experimental evidence suggest a second function of ABCA1 in cellular cholesterol homeostasis in addition to mediating cholesterol efflux. Here, we report the unexpected finding that ABCA1 also plays a key role in facilitating retrograde sterol transport from the PM to the ER for sterol sensing. Deficiency in ABCA1 delays sterol esterification at the ER and activates the SREBP-2 cleavage pathway. The intrinsic ATPase activity in ABCA1 is required to facilitate retrograde sterol transport. ABCA1 deficiency causes alternation of PM composition and hampers a clathrin-independent endocytic activity that is required for ER sterol sensing. Our finding identifies ABCA1 as a key macromolecule facilitating bidirectional sterol movement at the PM and shows that ABCA1 controls retrograde sterol transport by modulating a certain clathrin-independent endocytic process.


Journal of the American Heart Association | 2013

Medial and Intimal Calcification in Chronic Kidney Disease: Stressing the Contributions

Maximillian A. Rogers; Claudia Goettsch; Elena Aikawa

Cardiovascular calcification is a prominent feature of chronic inflammatory disorders that associate with significant morbidity and mortality. Vascular calcification is highly prevalent in patients with chronic kidney disease (CKD), a multifactorial disorder. CKD often results from hypertension and diabetes, and patients with CKD are among the highest-risk groups for cardiovascular events. Notably, CKD accelerates the development of atherosclerosis. Our group and others have demonstrated that CKD causes excessive vascular inflammation and calcification. 1 CKD is characterized


Circulation | 2015

A Not-So-Little Role for Lipoprotein(a) in the Development of Calcific Aortic Valve Disease

Maximillian A. Rogers; Elena Aikawa

Alterations in lipid metabolism and inflammatory processes are well established as potential risk factors in the development and progression of cardiovascular disease.1 However, with complications ranging from valve dysfunction to arrhythmia to myocardial infarction and stroke, the underlying mechanisms may be as varied as cardiovascular disease itself. On the other hand, the reoccurrence of common molecular and cellular pathways identified in the collective body of cardiovascular research could suggest shared initiators or mechanistic nodes between seemingly divergent processes, including lipid metabolism and inflammation. One area where this may hold true is cardiovascular calcification, in which dysregulated mineral metabolism in cardiovascular tissues leads to increased morbidity and mortality. Article see p 677 Calcification of soft tissues results from the deposition of calcium, largely in the form of hydroxyapatite, in the vascular wall or valve leaflets. Previously thought to be a passive degenerative process, it has become increasingly apparent that cardiovascular calcification is an active process initiated by many triggers. Recent studies have demonstrated variation in the LPA gene, which determines the plasma concentration of lipoprotein(a) [Lp(a); pronounced “L P little a”] to be associated with calcific aortic valve disease (CAVD).2,3 Lp(a) consists of a low-density lipoprotein (LDL)–like particle in which apolipoprotein(a) is covalently bound to apolipoprotein B. Additionally, Lp(a) is a genetic risk factor for atherosclerotic events.4 As in atherosclerosis, calcifications in CAVD localize to areas with lipoprotein accumulation and inflammatory cell infiltration, suggesting a shared disease process.5 However, some noticeable differences exist, including increased mechanical stresses and calcification-involved valve obstruction in CAVD as opposed to microcalcifications leading atherosclerosis plaque rupture.6 In this issue of Circulation , Bouchareb et al7 propose a highly plausible mechanistic pathway through which Lp(a) and valve interstitial cell (VIC)–derived autotaxin may induce valve calcification by regulating inflammation-induced bone …


Journal of Biological Chemistry | 2016

Myeloid Acyl-CoA:Cholesterol Acyltransferase 1 Deficiency Reduces Lesion Macrophage Content and Suppresses Atherosclerosis Progression

Li-Hao Huang; Elaina Melton; Haibo Li; Paul Sohn; Maximillian A. Rogers; Mary Jo Mulligan-Kehoe; Steven Fiering; William F. Hickey; Catherine C. Y. Chang; Ta-Yuan Chang

Acyl-CoA:cholesterol acyltransferase 1 (Acat1) converts cellular cholesterol to cholesteryl esters and is considered a drug target for treating atherosclerosis. However, in mouse models for atherosclerosis, global Acat1 knockout (Acat1−/−) did not prevent lesion development. Acat1−/− increased apoptosis within lesions and led to several additional undesirable phenotypes, including hair loss, dry eye, leukocytosis, xanthomatosis, and a reduced life span. To determine the roles of Acat1 in monocytes/macrophages in atherosclerosis, we produced a myeloid-specific Acat1 knockout (Acat1−M/−M) mouse and showed that, in the Apoe knockout (Apoe−/−) mouse model for atherosclerosis, Acat1−M/−M decreased the plaque area and reduced lesion size without causing leukocytosis, dry eye, hair loss, or a reduced life span. Acat1−M/−M enhanced xanthomatosis in apoe−/− mice, a skin disease that is not associated with diet-induced atherosclerosis in humans. Analyses of atherosclerotic lesions showed that Acat1−M/−M reduced macrophage numbers and diminished the cholesterol and cholesteryl ester load without causing detectable apoptotic cell death. Leukocyte migration analysis in vivo showed that Acat1−M/−M caused much fewer leukocytes to appear at the activated endothelium. Studies in inflammatory (Ly6Chi-positive) monocytes and in cultured macrophages showed that inhibiting ACAT1 by gene knockout or by pharmacological inhibition caused a significant decrease in integrin β 1 (CD29) expression in activated monocytes/macrophages. The sparse presence of lesion macrophages without Acat1 can therefore, in part, be attributed to decreased interaction between inflammatory monocytes/macrophages lacking Acat1 and the activated endothelium. We conclude that targeting ACAT1 in a myeloid cell lineage suppresses atherosclerosis progression while avoiding many of the undesirable side effects caused by global Acat1 inhibition.


Journal of Biological Chemistry | 2012

Cellular Pregnenolone Esterification by Acyl-CoA:Cholesterol Acyltransferase

Maximillian A. Rogers; Jay Liu; Elena Y. Bryleva; Alan L. Rockwood; A. Wayne Meikle; David J. Shapiro; Boris Vaisman; Alan T. Remaley; Catherine C. Y. Chang; Ta-Yuan Chang

Background: Various sterols serve as substrates and allosteric activators for ACAT. Results: Pregnelonone is the only natural sterol that is an ACAT substrate but not an ACAT activator. Conclusion: ACAT, along with LCAT contributes to control cellular pregnenolone ester content. Significance: This work provides a new function for ACAT. Pregnenolone (PREG) can be converted to PREG esters (PE) by the plasma enzyme lecithin: cholesterol acyltransferase (LCAT), and by other enzyme(s) with unknown identity. Acyl-CoA:cholesterol acyltransferase 1 and 2 (ACAT1 and ACAT2) convert various sterols to steryl esters; their activities are activated by cholesterol. PREG is a sterol-like molecule, with 3-β-hydroxy moiety at steroid ring A, but with much shorter side chain at steroid ring D. Here we show that without cholesterol, PREG is a poor ACAT substrate; with cholesterol, the Vmax for PREG esterification increases by 100-fold. The binding affinity of ACAT1 for PREG is 30–50-fold stronger than that for cholesterol; however, PREG is only a substrate but not an activator, while cholesterol is both a substrate and an activator. These results indicate that the sterol substrate site in ACAT1 does not involve significant sterol-phospholipid interaction, while the sterol activator site does. Studies utilizing small molecule ACAT inhibitors show that ACAT plays a key role in PREG esterification in various cell types examined. Mice lacking ACAT1 or ACAT2 do not have decreased PREG ester contents in adrenals, nor do they have altered levels of the three major secreted adrenal steroids in serum. Mice lacking LCAT have decreased levels of PREG esters in the adrenals. These results suggest LCAT along with ACAT1/ACAT2 contribute to control pregnenolone ester content in different cell types and tissues.


Iubmb Life | 2010

Neuronal cholesterol esterification by ACAT1 in Alzheimer's disease.

Ta-Yuan Chang; Catherine C. Y. Chang; Elena Y. Bryleva; Maximillian A. Rogers; Stephanie R. Murphy

Cholesterol has been implicated in various neurodegenerative diseases. Here we review the connection between cholesterol and Alzheimers disease (AD), focusing on a recent study that links neuronal cholesterol esterification with biosynthesis of 24(S)‐hydroxycholesterol and the fate of human amyloid precursor protein in a mouse model of AD. We also briefly evaluate the potential of ACAT1 as a drug target for AD.


Circulation Research | 2017

Dynamin-Related Protein 1 Inhibition Attenuates Cardiovascular Calcification in the Presence of Oxidative Stress

Maximillian A. Rogers; Natalia Maldonado; Joshua D. Hutcheson; Claudia Goettsch; Shinji Goto; Iwao Yamada; Tyler Faits; Hiromi Sesaki; Masanori Aikawa; Elena Aikawa

Rationale: Mitochondrial changes occur during cell differentiation and cardiovascular disease. DRP1 (dynamin-related protein 1) is a key regulator of mitochondrial fission. We hypothesized that DRP1 plays a role in cardiovascular calcification, a process involving cell differentiation and a major clinical problem with high unmet needs. Objective: To examine the effects of osteogenic promoting conditions on DRP1 and whether DRP1 inhibition alters the development of cardiovascular calcification. Methods and Results: DRP1 was enriched in calcified regions of human carotid arteries, examined by immunohistochemistry. Osteogenic differentiation of primary human vascular smooth muscle cells increased DRP1 expression. DRP1 inhibition in human smooth muscle cells undergoing osteogenic differentiation attenuated matrix mineralization, cytoskeletal rearrangement, mitochondrial dysfunction, and reduced type 1 collagen secretion and alkaline phosphatase activity. DRP1 protein was observed in calcified human aortic valves, and DRP1 RNA interference reduced primary human valve interstitial cell calcification. Mice heterozygous for Drp1 deletion did not exhibit altered vascular pathology in a proprotein convertase subtilisin/kexin type 9 gain-of-function atherosclerosis model. However, when mineralization was induced via oxidative stress, DRP1 inhibition attenuated mouse and human smooth muscle cell calcification. Femur bone density was unchanged in mice heterozygous for Drp1 deletion, and DRP1 inhibition attenuated oxidative stress–mediated dysfunction in human bone osteoblasts. Conclusions: We demonstrate a new function of DRP1 in regulating collagen secretion and cardiovascular calcification, a novel area of exploration for the potential development of new therapies to modify cellular fibrocalcific response in cardiovascular diseases. Our data also support a role of mitochondrial dynamics in regulating oxidative stress–mediated arterial calcium accrual and bone loss.

Collaboration


Dive into the Maximillian A. Rogers's collaboration.

Top Co-Authors

Avatar

Elena Aikawa

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masanori Aikawa

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claudia Goettsch

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Joshua D. Hutcheson

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tan Pham

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Sumihiko Hagita

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Andrew K. Mlynarchik

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge