Network


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

Hotspot


Dive into the research topics where Hilaire C. Lam is active.

Publication


Featured researches published by Hilaire C. Lam.


Nature Immunology | 2011

Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome

Kiichi Nakahira; Jeffrey A. Haspel; Vijay A. K. Rathinam; Seon-Jin Lee; Tamas Dolinay; Hilaire C. Lam; Joshua A. Englert; Marlene Rabinovitch; Manuela Cernadas; Hong Pyo Kim; Katherine A. Fitzgerald; Stefan W. Ryter; Augustine M. K. Choi

Autophagy, a cellular process for organelle and protein turnover, regulates innate immune responses. Here we demonstrate that depletion of the autophagic proteins LC3B and beclin 1 enhanced the activation of caspase-1 and secretion of interleukin 1β (IL-1β) and IL-18. Depletion of autophagic proteins promoted the accumulation of dysfunctional mitochondria and cytosolic translocation of mitochondrial DNA (mtDNA) in response to lipopolysaccharide (LPS) and ATP in macrophages. Release of mtDNA into the cytosol depended on the NALP3 inflammasome and mitochondrial reactive oxygen species (ROS). Cytosolic mtDNA contributed to the secretion of IL-1β and IL-18 in response to LPS and ATP. LC3B-deficient mice produced more caspase-1-dependent cytokines in two sepsis models and were susceptible to LPS-induced mortality. Our study suggests that autophagic proteins regulate NALP3-dependent inflammation by preserving mitochondrial integrity.


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

Autophagy protein microtubule-associated protein 1 light chain-3B (LC3B) activates extrinsic apoptosis during cigarette smoke-induced emphysema

Zhihua Chen; Hilaire C. Lam; Yang Jin; Hong-Pyo Kim; Jiao-Fei Cao; Seon-Jin Lee; Emeka Ifedigbo; Harikrishnan Parameswaran; Stefan W. Ryter; Augustine M. K. Choi

Chronic obstructive pulmonary disease (COPD) is a debilitating disease caused by chronic exposure to cigarette smoke (CS), which involves airway obstruction and alveolar loss (i.e., emphysema). The mechanisms of COPD pathogenesis remain unclear. Our previous studies demonstrated elevated autophagy in human COPD lung, and as a cellular and tissue response to CS exposure in an experimental model of emphysema in vivo. We identified the autophagic protein microtubule-associated protein 1 light chain-3B (LC3B) as a positive regulator of CS-induced lung epithelial cell death. We now extend these initial observations to explore the mechanism by which LC3B mediates CS-induced apoptosis and emphysema development in vivo. Here, we observed that LC3B−/− mice had significantly decreased levels of apoptosis in the lungs after CS exposure, and displayed resistance to CS-induced airspace enlargement, relative to WT littermate mice. We found that LC3B associated with the extrinsic apoptotic factor Fas in lipid rafts in an interaction mediated by caveolin-1 (Cav-1). The siRNA-dependent knockdown of Cav-1 sensitized epithelial cells to CS-induced apoptosis, as evidenced by enhanced death-inducing signaling complex formation and caspase activation. Furthermore, Cav-1−/− mice exhibited higher levels of autophagy and apoptosis in the lung in response to chronic CS exposure in vivo. In conclusion, we demonstrate a pivotal role for the autophagic protein LC3B in CS-induced apoptosis and emphysema, suggestive of novel therapeutic targets for COPD treatment. This study also introduces a mechanism by which LC3B, through interactions with Cav-1 and Fas, can regulate apoptosis.


Journal of Clinical Investigation | 2013

Histone deacetylase 6–mediated selective autophagy regulates COPD-associated cilia dysfunction

Hilaire C. Lam; Suzanne M. Cloonan; Abhiram R. Bhashyam; Jeffery A. Haspel; Anju Singh; J. Fah Sathirapongsasuti; Morgan Cervo; Hongwei Yao; Anna L. Chung; Kenji Mizumura; Chang Hyeok An; Bin Shan; Jonathan Franks; Kathleen J. Haley; Caroline A. Owen; Yohannes Tesfaigzi; George R. Washko; John Quackenbush; Edwin K. Silverman; Irfan Rahman; Hong Pyo Kim; Ashfaq Mahmood; Shyam Biswal; Stefan W. Ryter; Augustine M. K. Choi

Chronic obstructive pulmonary disease (COPD) involves aberrant airway inflammatory responses to cigarette smoke (CS) that are associated with epithelial cell dysfunction, cilia shortening, and mucociliary clearance disruption. Exposure to CS reduced cilia length and induced autophagy in vivo and in differentiated mouse tracheal epithelial cells (MTECs). Autophagy-impaired (Becn1+/- or Map1lc3B-/-) mice and MTECs resisted CS-induced cilia shortening. Furthermore, CS increased the autophagic turnover of ciliary proteins, indicating that autophagy may regulate cilia homeostasis. We identified cytosolic deacetylase HDAC6 as a critical regulator of autophagy-mediated cilia shortening during CS exposure. Mice bearing an X chromosome deletion of Hdac6 (Hdac6-/Y) and MTECs from these mice had reduced autophagy and were protected from CS-induced cilia shortening. Autophagy-impaired Becn1-/-, Map1lc3B-/-, and Hdac6-/Y mice or mice injected with an HDAC6 inhibitor were protected from CS-induced mucociliary clearance (MCC) disruption. MCC was preserved in mice given the chemical chaperone 4-phenylbutyric acid, but was disrupted in mice lacking the transcription factor NRF2, suggesting that oxidative stress and altered proteostasis contribute to the disruption of MCC. Analysis of human COPD specimens revealed epigenetic deregulation of HDAC6 by hypomethylation and increased protein expression in the airways. We conclude that an autophagy-dependent pathway regulates cilia length during CS exposure and has potential as a therapeutic target for COPD.


Journal of Experimental Medicine | 2014

Regulation of YAP by mTOR and autophagy reveals a therapeutic target of tuberous sclerosis complex

Ning Liang; Chi Zhang; Patricia Dill; Ganna Panasyuk; Delphine Pion; Vonda Koka; Morgan Gallazzini; Eric N. Olson; Hilaire C. Lam; Elizabeth P. Henske; Zheng Dong; Udayan Apte; Nicolas Pallet; Randy L. Johnson; Fabiola Terzi; David J. Kwiatkowski; Jean-Yves Scoazec; Guido Martignoni; Mario Pende

Liang et al. find that the tumor suppressors TSC1 and TSC2, defects in which underlie the genetic disease Tuberous Sclerosis Complex (TSC), drive the mTOR-dependent autophagosomal destruction of the transcriptional activator YAP. Blocking YAP inhibited the abnormal proliferation of TSC1/2-deficient human cells and reversed TSC-like disease symptoms in mosaic Tsc1 mutant mice.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2012

TLR4 deficiency promotes autophagy during cigarette smoke-induced pulmonary emphysema

Chang Hyeok An; Xiao Mei Wang; Hilaire C. Lam; Emeka Ifedigbo; George R. Washko; Stefan W. Ryter; Augustine M. K. Choi

Toll-like receptors (TLRs) exert important nonimmune functions in lung homeostasis. TLR4 deficiency promotes pulmonary emphysema. We examined the role of TLR4 in regulating cigarette smoke (CS)-induced autophagy, apoptosis, and emphysema. Lung tissue was obtained from chronic obstructive lung disease (COPD) patients. C3H/HeJ (Tlr4-mutated) mice and C57BL/10ScNJ (Tlr4-deficient) mice and their respective control strains were exposed to chronic CS or air. Human or mouse epithelial cells (wild-type, Tlr4-knockdown, and Tlr4-deficient) were exposed to CS-extract (CSE). Samples were analyzed for TLR4 expression, and for autophagic or apoptotic proteins by Western blot analysis or confocal imaging. Chronic obstructive lung disease lung tissues and human pulmonary epithelial cells exposed to CSE displayed increased TLR4 expression, and increased autophagic [microtubule-associated protein-1 light-chain-3B (LC3B)] and apoptotic (cleaved caspase-3) markers. Beas-2B cells transfected with TLR4 siRNA displayed increased expression of LC3B relative to control cells, basally and after exposure to CSE. The basal and CSE-inducible expression of LC3B and cleaved caspase-3 were elevated in pulmonary alveolar type II cells from Tlr4-deficient mice. Wild-type mice subjected to chronic CS-exposure displayed airspace enlargement;, however, the Tlr4-mutated or Tlr4-deficient mice exhibited a marked increase in airspace relative to wild-type mice after CS-exposure. The Tlr4-mutated or Tlr4-deficient mice showed higher levels of LC3B under basal conditions and after CS exposure. The expression of cleaved caspase-3 was markedly increased in Tlr4-deficient mice exposed to CS. We describe a protective regulatory function of TLR4 against emphysematous changes of the lung in response to CS.


Autophagy | 2014

“Ciliophagy”: The consumption of cilia components by autophagy

Suzanne M. Cloonan; Hilaire C. Lam; Stefan W. Ryter; Augustine M. K. Choi

Chronic obstructive pulmonary disease (COPD) involves aberrant airway inflammatory responses to cigarette smoke (CS) associated with respiratory epithelial cell cilia shortening and impaired mucociliary clearance (MCC). The underlying cellular and molecular mechanisms for CS-associated cilia shortening have remained incompletely understood. We have previously demonstrated increased autophagy in the lungs of COPD patients; however, whether or not this process is selective for specific autophagic targets in the lung was not elucidated. Based on observations that increased morphological and biochemical indicators of autophagy correlate with cilia shortening in our models, we posited that autophagy might regulate cilia length in response to CS in the lung. We demonstrate that CS-induced cilia shortening occurs through an autophagy-dependent mechanism mediated by the deacetylase HDAC6 (histone deacetylase 6). Autophagy-impaired (Becn1(+/-), map1lc3b(-/-), or Hdac6(-/Y)) mice resist CS-induced cilia shortening. Furthermore, cilia components are identified as autophagic substrates during CS exposure. Assessment of airway cilia function using a 3D MCC assay demonstrates that Becn1(+/-), map1lc3b(-/-), and Hdac6(-/Y) mice or mice injected with the HDAC6 inhibitor tubastatin A are protected from CS-associated mucociliary dysfunction. We concluded that an autophagy-dependent pathway regulates cilia length during CS exposure, which identifies new pathways and targets in COPD.Chronic obstructive pulmonary disease (COPD) involves aberrant airway inflammatory responses to cigarette smoke (CS) associated with respiratory epithelial cell cilia shortening and impaired mucociliary clearance (MCC). The underlying cellular and molecular mechanisms for CS-associated cilia shortening have remained incompletely understood. We have previously demonstrated increased autophagy in the lungs of COPD patients; however, whether or not this process is selective for specific autophagic targets in the lung was not elucidated. Based on observations that increased morphological and biochemical indicators of autophagy correlate with cilia shortening in our models, we posited that autophagy might regulate cilia length in response to CS in the lung. We demonstrate that CS-induced cilia shortening occurs through an autophagy-dependent mechanism mediated by the deacetylase HDAC6 (histone deacetylase 6). Autophagy-impaired (Becn1+/−, map1lc3b−/−, or Hdac6-/Y) mice resist CS-induced cilia shortening. Furthermore, cilia components are identified as autophagic substrates during CS exposure. Assessment of airway cilia function using a 3D MCC assay demonstrates that Becn1+/−, map1lc3b−/−, and Hdac6-/Y mice or mice injected with the HDAC6 inhibitor tubastatin A are protected from CS-associated mucociliary dysfunction. We concluded that an autophagy-dependent pathway regulates cilia length during CS exposure, which identifies new pathways and targets in COPD.


Autophagy | 2011

Deadly triplex: Smoke, autophagy and apoptosis

Stefan W. Ryter; Hilaire C. Lam; Zhihua Chen; Augustine M. K. Choi

Autophagy, a cellular program for organelle and protein turnover, represents primarily a cell survival mechanism. However, the role of autophagy in the regulation of apoptosis remains unclear. We have observed increases in morphological and biochemical indicators of autophagy in human lung from patients with chronic obstructive pulmonary disease (COPD). Furthermore, we observed induction of autophagic markers in mouse lung subjected to chronic cigarette smoke exposure. Recently, we investigated the role of the autophagic protein microtubule-associated protein 1 light chain 3B (LC3B) as a regulator of lung cell death. We found that LC3B knockout (LC3B-/-) mice subjected to chronic cigarette smoke exposure have reduced lung apoptosis, and resist airspace enlargement, relative to wild-type mice. We therefore examined the mechanisms by which LC3B can regulate apoptosis in epithelial cells. We found that LC3B forms a complex with the death receptor Fas in lipid rafts of epithelial cells, which requires the caveolae-resident protein caveolin-1. Genetic interference of caveolin-1 in epithelial cells augments cigarette smoke-induced apoptosis. Caveolin-1 knockout mice exhibit increased autophagic markers, apoptosis, and airspace enlargement in the lung in response to chronic cigarette smoke. These studies demonstrate that LC3B can promote tissue injury during chronic cigarette smoke exposure, and suggest a mechanism by which LC3B, through interactions with caveolin-1 and Fas, can regulate apoptosis. Targeting the autophagic pathway may represent an experimental therapeutic strategy when designing new approaches to COPD treatment.


Journal of Visualized Experiments | 2011

Isolation of mouse respiratory epithelial cells and exposure to experimental cigarette smoke at air liquid interface.

Hilaire C. Lam; Augustine M. K. Choi; Stefan W. Ryter

Pulmonary epithelial cells can be isolated from the respiratory tract of mice and cultured at air-liquid interface (ALI) as a model of differentiated respiratory epithelium. A protocol is described for isolating and exposing these cells to mainstream cigarette smoke (CS), in order to study epithelial cell responses to CS exposure. The protocol consists of three parts: the isolation of airway epithelial cells from mouse trachea, the culturing of these cells at air-liquid interface (ALI) as fully differentiated epithelial cells, and the delivery of calibrated mainstream CS to these cells in culture. The ALI culture system allows the culture of respiratory epithelia under conditions that more closely resemble their physiological setting than ordinary liquid culture systems. The study of molecular and lung cellular responses to CS exposure is a critical component of understanding the impact of environmental air pollution on human health. Research findings in this area may ultimately contribute towards understanding the etiology of chronic obstructive pulmonary disease (COPD), and other tobacco-related diseases, which represent major global health problems.


Journal of Visualized Experiments | 2014

Cecal Ligation and Puncture-induced Sepsis as a Model To Study Autophagy in Mice

Ilias I. Siempos; Hilaire C. Lam; Yan Ding; Mary E. Choi; Augustine M. K. Choi; Stefan W. Ryter

Experimental sepsis can be induced in mice using the cecal ligation and puncture (CLP) method, which causes polymicrobial sepsis. Here, a protocol is provided to induce sepsis of varying severity in mice using the CLP technique. Autophagy is a fundamental tissue response to stress and pathogen invasion. Two current protocols to assess autophagy in vivo in the context of experimental sepsis are also presented here. (I) Transgenic mice expressing green fluorescence protein (GFP)-LC3 fusion protein are subjected to CLP. Localized enhancement of GFP signal (puncta), as assayed either by immunohistochemical or confocal assays, can be used to detect enhanced autophagosome formation and, thus, altered activation of the autophagy pathway. (II) Enhanced autophagic vacuole (autophagosome) formation per unit tissue area (as a marker of autophagy stimulation) can be quantified using electron microscopy. The study of autophagic responses to sepsis is a critical component of understanding the mechanisms by which tissues respond to infection. Research findings in this area may ultimately contribute towards understanding the pathogenesis of sepsis, which represents a major problem in critical care medicine.


Cancer Cell | 2017

mTORC1 Couples Nucleotide Synthesis to Nucleotide Demand Resulting in a Targetable Metabolic Vulnerability

Alexander J. Valvezan; Marc Turner; Amine Belaid; Hilaire C. Lam; Spencer K. Miller; Molly C. McNamara; Christian V. Baglini; Benjamin E. Housden; Norbert Perrimon; David J. Kwiatkowski; John M. Asara; Elizabeth P. Henske; Brendan D. Manning

The mechanistic target of rapamycin complex 1 (mTORC1) supports proliferation through parallel induction of key anabolic processes, including protein, lipid, and nucleotide synthesis. We hypothesized that these processes are coupled to maintain anabolic balance in cells with mTORC1 activation, a common event in human cancers. Loss of the tuberous sclerosis complex (TSC) tumor suppressors results in activation of mTORC1 and development of the tumor syndrome TSC. We find that pharmacological inhibitors of guanylate nucleotide synthesis have selective deleterious effects on TSC-deficient cells, including in mouse tumor models. This effect stems from replication stress and DNA damage caused by mTORC1-driven rRNA synthesis, which renders nucleotide pools limiting. These findings reveal a metabolic vulnerability downstream of mTORC1 triggered by anabolic imbalance.

Collaboration


Dive into the Hilaire C. Lam's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elizabeth P. Henske

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong Pyo Kim

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Harilaos Filippakis

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Nicola Alesi

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge