Network


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

Hotspot


Dive into the research topics where Babak Razani is active.

Publication


Featured researches published by Babak Razani.


Pharmacological Reviews | 2002

Caveolae: From Cell Biology to Animal Physiology

Babak Razani; Scott E. Woodman; Michael P. Lisanti

Among the membrane compartments of a cell, vesicles known as “caveolae” have long defied functional characterization. However, since the identification of a family of proteins termed “caveolins”, that form and reside in caveolae, a better understanding has emerged. It is now clear that caveolae do not merely play a singular role in the cell, but are pleiotropic in nature—serving to modulate many cellular functions. The purpose of this review is to explicate what is known about caveolins/caveolae and highlight growing areas of caveolar research.


Cell | 2001

Emerging Themes in Lipid Rafts and Caveolae

Ferruccio Galbiati; Babak Razani; Michael P. Lisanti

We would especially like to thank the following meeting participants who generously contributed to the organization and writing of this meeting review: Miguel Alonso, Toyoshi Fujimoto, Jean Gruenberg, Hai-Tao He, Vaclav Horejsi, Akihiro Kusumi, Tony Magee, Fred Maxfield, Satyajit Mayor, Mark McNiven, Roger Morris, Robert Parton, Radu-Virgil Stan, Claudia Stuermer, and Gisou van der Goot. M.P.L would like to thank Dr. Federica Sotgia for her support and inspiration.M.P.L. is supported by grants from the National Institutes of Health (NIH), the Muscular Dystrophy Association (MDA), the American Heart Association (AHA), and the Komen Breast Cancer Foundation, as well as a Hirschl/Weil-Caulier Career Scientist Award. F.G. is the recipient of a Scientist Development Grant from the American Heart Association (AHA). B.R. is supported by a National Institutes of Health Medical Scientist Training Grant (T32-GM07288).


Immunity | 2014

Embryonic and Adult-Derived Resident Cardiac Macrophages Are Maintained through Distinct Mechanisms at Steady State and during Inflammation

Slava Epelman; Kory J. Lavine; Anna E. Beaudin; Dorothy K. Sojka; Javier A. Carrero; Boris Calderon; Thaddeus Brija; Emmanuel L. Gautier; Stoyan Ivanov; Ansuman T. Satpathy; Joel D. Schilling; Reto A. Schwendener; Ismail Sergin; Babak Razani; E. Camilla Forsberg; Wayne M. Yokoyama; Emil R. Unanue; Marco Colonna; Gwendalyn J. Randolph; Douglas L. Mann

Cardiac macrophages are crucial for tissue repair after cardiac injury but are not well characterized. Here we identify four populations of cardiac macrophages. At steady state, resident macrophages were primarily maintained through local proliferation. However, after macrophage depletion or during cardiac inflammation, Ly6c(hi) monocytes contributed to all four macrophage populations, whereas resident macrophages also expanded numerically through proliferation. Genetic fate mapping revealed that yolk-sac and fetal monocyte progenitors gave rise to the majority of cardiac macrophages, and the heart was among a minority of organs in which substantial numbers of yolk-sac macrophages persisted in adulthood. CCR2 expression and dependence distinguished cardiac macrophages of adult monocyte versus embryonic origin. Transcriptional and functional data revealed that monocyte-derived macrophages coordinate cardiac inflammation, while playing redundant but lesser roles in antigen sampling and efferocytosis. These data highlight the presence of multiple cardiac macrophage subsets, with different functions, origins, and strategies to regulate compartment size.


Molecular and Cellular Biology | 2002

Caveolin-2-Deficient Mice Show Evidence of Severe Pulmonary Dysfunction without Disruption of Caveolae

Babak Razani; Xiao Bo Wang; Jeffery A. Engelman; Michela Battista; Guy Lagaud; Xiao Lan Zhang; Burkhard Kneitz; Harry Hou; George J. Christ; Winfried Edelmann; Michael P. Lisanti

ABSTRACT Caveolin-2 is a member of the caveolin gene family with no known function. Although caveolin-2 is coexpressed and heterooligomerizes with caveolin-1 in many cell types (most notably adipocytes and endothelial cells), caveolin-2 has traditionally been considered the dispensable structural partner of the widely studied caveolin-1. We now directly address the functional significance of caveolin-2 by genetically targeting the caveolin-2 locus (Cav-2) in mice. In the absence of caveolin-2 protein expression, caveolae still form and caveolin-1 maintains its localization in plasma membrane caveolae, although in certain tissues caveolin-1 is partially destabilized and shows modestly diminished protein levels. Despite an intact caveolar membrane system, the Cav-2-null lung parenchyma shows hypercellularity, with thickened alveolar septa and an increase in the number of endothelial cells. As a result of these pathological changes, these Cav-2-null mice are markedly exercise intolerant. Interestingly, these Cav-2-null phenotypes are identical to the ones we and others have recently reported for Cav-1-null mice. As caveolin-2 expression is also severely reduced in Cav-1-null mice, we conclude that caveolin-2 deficiency is the clear culprit in this lung disorder. Our analysis of several different phenotypes observed in caveolin-1-deficient mice (i.e., abnormal vascular responses and altered lipid homeostasis) reveals that Cav-2-null mice do not show any of these other phenotypes, indicating a selective role for caveolin-2 in lung function. Taken together, our data show for the first time a specific role for caveolin-2 in mammalian physiology independent of caveolin-1.


Cell Metabolism | 2012

Autophagy Links Inflammasomes to Atherosclerotic Progression

Babak Razani; Chu Feng; Trey Coleman; Roy Emanuel; Haitao Wen; Seungmin Hwang; Jenny P.-Y. Ting; Herbert W. Virgin; Michael B. Kastan; Clay F. Semenkovich

We investigated the role of autophagy in atherosclerosis. During plaque formation in mice, autophagic markers colocalized predominantly with macrophages (mφ). Atherosclerotic aortas had elevated levels of p62, suggesting that dysfunctional autophagy is characteristic of plaques. To determine whether autophagy directly influences atherogenesis, we characterized Beclin-1 heterozygous-null and mφ-specific ATG5-null (ATG5-mφKO) mice, commonly used models of autophagy haploinsufficiency and deficiency, respectively. Haploinsufficent Beclin-1 mice had no atherosclerotic phenotype, but ATG5-mφKO mice had increased plaques, suggesting an essential role for basal levels of autophagy in atheroprotection. Defective autophagy is associated with proatherogenic inflammasome activation. Classic inflammasome markers were robustly induced in ATG5-null mφ, especially when coincubated with cholesterol crystals. Moreover, cholesterol crystals appear to be increased in ATG5-mφKO plaques, suggesting a potentially vicious cycle of crystal formation and inflammasome activation in autophagy-deficient plaques. These results show that autophagy becomes dysfunctional in atherosclerosis and its deficiency promotes atherosclerosis in part through inflammasome hyperactivation.


American Journal of Pathology | 2002

Caveolin-1/3 Double-Knockout Mice Are Viable, but Lack Both Muscle and Non-Muscle Caveolae, and Develop a Severe Cardiomyopathic Phenotype

David S. Park; Scott E. Woodman; William Schubert; Alex W. Cohen; Philippe G. Frank; Madhulika Chandra; Jamshid Shirani; Babak Razani; Baiyu Tang; Linda A. Jelicks; Stephen M. Factor; Louis M. Weiss; Herbert B. Tanowitz; Michael P. Lisanti

The caveolin gene family consists of caveolins 1, 2, and 3. Caveolins 1 and 2 are co-expressed in many cell types, such as endothelial cells, fibroblasts, smooth muscle cells and adipocytes, where they form a heteroligomeric complex. In contrast, the expression of caveolin-3 is muscle-specific. Thus, the expression of caveolin-1 is required for caveolae formation in non-muscle cells, while the expression of caveolin-3 drives caveolae formation in striated muscle cell types (cardiac and skeletal). To create a truly caveolae-deficient mouse, we interbred Cav-1 null mice and Cav-3 null mice to generate Cav-1/Cav-3 double-knockout (Cav-1/3 dKO) mice. Here, we report that Cav-1/3 dKO mice are viable and fertile, despite the fact that they lack morphologically identifiable caveolae in endothelia, adipocytes, smooth muscle cells, skeletal muscle fibers, and cardiac myocytes. We also show that these mice are deficient in all three caveolin gene products, as caveolin-2 is unstable in the absence of caveolin-1. Interestingly, Cav-1/3 dKO mice develop a severe cardiomyopathy. At 2 months of age, analysis of Cav-1/3 dKO hearts via gated magnetic resonance imaging reveals a dramatic increase in left ventricular wall thickness, as compared with Cav-1-KO, Cav-3 KO, and wild-type mice. Further functional analysis of Cav-1/3 dKO hearts via transthoracic echocardiography demonstrates hypertrophy and dilation of the left ventricle, with a significant decrease in fractional shortening. As predicted, Northern analysis of RNA derived from the left ventricle of Cav-1/3 dKO mice shows a dramatic up-regulation of the atrial natriuretic factor message, a well-established biochemical marker of cardiac hypertrophy. Finally, histological analysis of Cav-1/3 dKO hearts reveals hypertrophy, disorganization, and degeneration of the cardiac myocytes, as well as chronic interstitial fibrosis and inflammation. Thus, dual ablation of both Cav-1 and Cav-3 genes in mice leads to a pleiotropic defect in caveolae formation and severe cardiomyopathy.


Journal of Clinical Investigation | 2001

Caveolin-deficient mice: insights into caveolar function human disease

Babak Razani; Michael P. Lisanti

In the past several decades, the discovery and detailed characterization of the various subcellular organelles has revealed the intricate workings of the cell. It is of great interest then, that one of the most prominent structures at the plasma membrane, the caveola, has remained the most enigmatic organelle found in cells. Together with their marker proteins, the caveolins, caveolae are present in large numbers in a variety of cell types. Although they were initially proposed to act as mere conduits for cellular uptake, akin to clathrin-coated vesicles, it is now clear that caveolae and caveolins have pleiotropic functions that modulate numerous cellular processes. Many proposed physiological roles for caveolae are controversial, making a rigorous analysis of their function imperative. A giant leap in this regard is the recent series of reports on the phenotypic characterization of caveolin-deficient mice. As loss of caveolin expression causes a concomitant loss in morphologically identifiable caveolae, such animals allow for the first definitive studies of the role of these organelles in various cells in vivo. In this review, we will discuss the known and proposed functions of caveolae in the context of these caveolin-deficient animal models, give a synopsis of the rapidly emerging themes in the field, and discuss the relevance of caveolae to the understanding of human disease.


American Journal of Pathology | 2002

Caveolin-1 Mutations (P132L and Null) and the Pathogenesis of Breast Cancer: Caveolin-1 (P132L) Behaves in a Dominant-Negative Manner and Caveolin-1 (−/−) Null Mice Show Mammary Epithelial Cell Hyperplasia

Hyangkyu Lee; David S. Park; Babak Razani; Robert G. Russell; Richard G. Pestell; Michael P. Lisanti

Caveolin-1 (Cav-1) is the principal structural protein of caveolae membranes that are found in most cells types, including mammary epithelial cells. Recently, we mapped the human CAV1 gene to a suspected tumor suppressor locus (7q31.1/D7S522) that is deleted in a variety of human cancers, as well as mammary tumors. In addition, the CAV1 gene is mutated (P132L) in up to approximately 16% of human breast cancers. The mechanism by which deletion or mutation of the Cav-1 gene contributes to mammary tumorigenesis remains unknown. To understand the role of the Cav-1 (P132L) mutation in the pathogenesis of human breast cancers, we generated the same mutation in wild-type (WT) Cav-1 and studied its behavior in cultured cells. Interestingly, the P132L mutation leads to formation of misfolded Cav-1 oligomers that are retained within the Golgi complex and are not targeted to caveolae or the plasma membrane. To examine whether the Cav-1 (P132L) mutant behaves in a dominant-negative manner, we next co-transfected cells with Cav-1 (P132L) and WT Cav-1, and evaluated their caveolar targeting. Our results indicate that Cav-1 (P132L) behaves in a dominant-negative manner, causing the mislocalization and intracellular retention of WT Cav-1. Virtually identical results were obtained when Cav-1 (P132L) was stably expressed at physiological levels in a nontransformed human mammary epithelial cell line (hTERT-HME1). These data provide a molecular explanation for why only a single mutated CAV1 allele is found in patients with breast cancer. Thus, we next investigated if functional inactivation of Cav-1 gene expression leads to mammary tumorigenesis in vivo. For this purpose, we performed mammary gland analysis on Cav-1-deficient mice (-/-) that harbor a targeted disruption of the Cav-1 gene (a null mutation). Interestingly, we show that inactivation of Cav-1 gene expression leads to mammary epithelial cell hyperplasia, even in 6-week-old virgin female mice. These data clearly implicate loss of functional Cav-1 in the pathogenesis of mammary epithelial cell hyperplasia, and suggest that Cav-1-null mice represent a novel animal model to study premalignant mammary disease.


Journal of Biological Chemistry | 1999

Regulation of cAMP-mediated Signal Transduction via Interaction of Caveolins with the Catalytic Subunit of Protein Kinase A

Babak Razani; Charles S. Rubin; Michael P. Lisanti

cAMP-dependent processes are essential for cell growth, differentiation, and homeostasis. The classic components of this system include the serpentine receptors, heterotrimeric G-proteins, adenylyl cyclase, protein kinase A (PKA), and numerous downstream target substrates. Evidence is accumulating that some members of this cascade are concentrated within membrane microdomains, termed caveolae and caveolae-related domains. In addition, the caveolin-1 protein has been shown to interact with some of these components, and this interaction inhibits their enzymatic activity. However, the functional effects of caveolins on cAMP-mediated signaling at the most pivotal step, PKA activation, remain unknown. Here, we show that caveolin-1 can dramatically inhibit cAMP-dependent signaling in vivo. We provide evidence for a direct interaction between caveolin-1 and the catalytic subunit of PKA both in vitro and in vivo. Caveolin-1 binding appears to be mediated both by the caveolin scaffolding domain (residues 82–101) and a portion of the C-terminal domain (residues 135–156). Further functional analysis indicates that caveolin-based peptides derived from these binding regions can inhibit the catalytic activity of purified PKA in vitro. Mutational analysis of the caveolin scaffolding domain reveals that a series of aromatic residues within the caveolin scaffolding domain are critical for mediating inhibition of PKA. In addition, co-expression of caveolin-1 and PKA in cultured cells results in their co-localization as seen by immunofluorescence microscopy. In cells co-expressing caveolin-1 and PKA, PKA assumed a punctate distribution that coincided with the distribution of caveolin-1. In contrast, in cells expressing PKA alone, PKA was localized throughout the cytoplasm and yielded a diffuse staining pattern. Taken together, our results suggest that the direct inhibition of PKA by caveolin-1 is an important and previously unrecognized mechanism for modulating cAMP-mediated signaling.


Journal of Biological Chemistry | 1999

Angiogenesis Activators and Inhibitors Differentially Regulate Caveolin-1 Expression and Caveolae Formation in Vascular Endothelial Cells ANGIOGENESIS INHIBITORS BLOCK VASCULAR ENDOTHELIAL GROWTH FACTOR-INDUCED DOWN-REGULATION OF CAVEOLIN-1

Jun Liu; Babak Razani; Shaoqing Tang; Bruce I. Terman; Ware Ja; Michael P. Lisanti

Angiogenesis is the process by which new blood vessels are formed via proliferation of vascular endothelial cells. A variety of angiogenesis inhibitors that antagonize the effects of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) have recently been identified. However, the mechanism by which these diverse angiogenesis inhibitors exert their common effects remains largely unknown. Caveolin-1 and -2 are known to be highly expressed in vascular endothelial cells both in vitro andin vivo. Here, we examine the potential role of caveolins in the angiogenic response. For this purpose, we used the well established human umbilical vein endothelial cell line, ECV 304. Treatment of ECV 304 cells with known angiogenic growth factors (VEGF, bFGF, or hepatocyte growth factor/scatter factor), resulted in a dramatic reduction in the expression of caveolin-1. This down-regulation event was selective for caveolin-1, as caveolin-2 levels remained constant under these conditions of growth factor stimulation. VEGF-induced down-regulation of caveolin-1 expression also resulted in the morphological loss of cell surface caveolae organelles as seen by transmission electron microscopy. A variety of well characterized angiogenesis inhibitors (including angiostatin, fumagillin, 2-methoxy estradiol, transforming growth factor-β, and thalidomide) effectively blocked VEGF-induced down-regulation of caveolin-1 as seen by immunoblotting and immunofluorescence microscopy. However, treatment with angiogenesis inhibitors alone did not significantly affect the expression of caveolin-1. PD98059, a specific inhibitor of mitogen-activated protein kinase and a known angiogenesis inhibitor, also blocked the observed VEGF-induced down-regulation of caveolin-1. Furthermore, we show that caveolin-1 can function as a negative regulator of VEGF-R (KDR) signal transduction in vivo. Thus, down-regulation of caveolin-1 may be an important step along the pathway toward endothelial cell proliferation.

Collaboration


Dive into the Babak Razani's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ismail Sergin

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Trent D. Evans

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Clay F. Semenkovich

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

David S. Park

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Richard G. Pestell

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Xiangyu Zhang

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Roy Emanuel

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Somashubhra Bhattacharya

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge